1
|
Siebert JR, Kennedy K, Osterhout DJ. Neurons Are Not All the Same: Diversity in Neuronal Populations and Their Intrinsic Responses to Spinal Cord Injury. ASN Neuro 2025; 17:2440299. [PMID: 39819292 DOI: 10.1080/17590914.2024.2440299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/19/2025] Open
Abstract
Functional recovery following spinal cord injury will require the regeneration and repair of damaged neuronal pathways. It is well known that the tissue response to injury involves inflammation and the formation of a glial scar at the lesion site, which significantly impairs the capacity for neuronal regeneration and functional recovery. There are initial attempts by both supraspinal and intraspinal neurons to regenerate damaged axons, often influenced by the neighboring tissue pathology. Many experimental therapeutic strategies are targeted to further stimulate the initial axonal regrowth, with little consideration for the diversity of the affected neuronal populations. Notably, recent studies reveal that the neuronal response to injury is variable, based on multiple factors, including the location of the injury with respect to the neuronal cell bodies and the affected neuronal populations. New insights into regenerative mechanisms have shown that neurons are not homogenous but instead exhibit a wide array of diversity in their gene expression, physiology, and intrinsic responses to injury. Understanding this diverse intrinsic response is crucial, as complete functional recovery requires the successful coordinated regeneration and reorganization of various neuron pathways.
Collapse
Affiliation(s)
- Justin R Siebert
- Physician Assistant Studies Program, Department of Health Care and Administration, Slippery Rock University of Pennsylvania, Slippery Rock, PA, USA
| | - Kiersten Kennedy
- Norton College of Medicine, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Donna J Osterhout
- Department of Cell & Developmental Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
2
|
Ebrahimi M, Ahmadieh H, Rezaei Kanavi M, Safi S, Alipour-Parsa S, Advani S, Sorenson CM, Sheibani N. Shared signaling pathways and comprehensive therapeutic approaches among diabetes complications. Front Med (Lausanne) 2025; 11:1497750. [PMID: 39845838 PMCID: PMC11750824 DOI: 10.3389/fmed.2024.1497750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/17/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
The growing global prevalence of diabetes mellitus (DM), along with its associated complications, continues to rise. When clinically detected most DM complications are irreversible. It is therefore crucial to detect and address these complications early and systematically in order to improve patient care and outcomes. The current clinical practice often prioritizes DM complications by addressing one complication while overlooking others that could occur. It is proposed that the commonly targeted cell types including vascular cells, immune cells, glial cells, and fibroblasts that mediate DM complications, might share early responses to diabetes. In addition, the impact of one complication could be influenced by other complications. Recognizing and focusing on the shared early responses among DM complications, and the impacted cellular constituents, will allow to simultaneously address all DM-related complications and limit adverse treatment impacts. This review explores the current understanding of shared pathological signaling mechanisms among DM complications and recognizes new concepts that will benefit from further investigation in both basic and clinical settings. The ultimate goal is to develop more comprehensive treatment strategies, which effectively impact DM complications in multiple organs and improve patient care and outcomes.
Collapse
Affiliation(s)
- Moein Ebrahimi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Ahmadieh
- Ophthalmic Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mozhgan Rezaei Kanavi
- Ocular Tissue Engineering Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sare Safi
- Ophthalmic Epidemiology Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Alipour-Parsa
- Cardiovascular Research Center, Shahid Modarres Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soroor Advani
- Neurology Department, Shohada Tajrish Hospital, Shahid-Beheshti University of Medical Sciences, Tehran, Iran
| | - Christine M. Sorenson
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Nader Sheibani
- Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| |
Collapse
|
3
|
Si Z, Fan Y, Wang M, Zhao J, Zhang Y, Liu D, Zheng Y. The role of RGC degeneration in the pathogenesis of glaucoma. Int J Biol Sci 2025; 21:211-232. [PMID: 39744428 PMCID: PMC11667808 DOI: 10.7150/ijbs.103222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/04/2024] [Accepted: 11/09/2024] [Indexed: 01/11/2025] Open
Abstract
Glaucoma is a neurodegenerative disorder marked by the loss of retinal ganglion cells (RGCs) and axonal degeneration, resulting in irreversible vision impairment. While intraocular pressure (IOP) is presently acknowledged as the sole modifiable risk factor, the sensitivity of RGCs to IOP varies among individuals. Consequently, progressive vision loss may ensue even when IOP is effectively managed. This review consolidates current knowledge regarding the pathogenesis of RGCs in glaucoma and various neurodegenerative diseases. It delves into the mechanisms underlying RGC transsynaptic degeneration and axonal defects in glaucoma from a pathophysiological standpoint, and it elucidates the alterations in the visual pathway throughout the progression of the disease. Furthermore, the article outlines neuroprotective and nerve regeneration strategies aimed at vision restoration in glaucoma patients, offers insights for clinical management of the condition, and investigates prospective avenues for gene therapy.
Collapse
Affiliation(s)
- Zhibo Si
- Department of Ophthalmology, The Second Hospital of Jilin University, Jilin University, Changchun 130000, Jilin, China
| | - Yuhang Fan
- Department of Geriatrics, The Second Hospital of Tianjin Medical University, Tianjin Medical University, Tianjin 300000, China
| | - Mingxuan Wang
- Department of Ophthalmology, The Second Hospital of Jilin University, Jilin University, Changchun 130000, Jilin, China
| | - Jing Zhao
- Department of Ophthalmology, The Second Hospital of Jilin University, Jilin University, Changchun 130000, Jilin, China
| | - Yunmei Zhang
- Department of Ophthalmology, Changchun Yishidun Eye Hospital, Changchun 130000, Jilin, China
| | - Dongmei Liu
- Department of Ophthalmology, Changchun Yishidun Eye Hospital, Changchun 130000, Jilin, China
| | - Yajuan Zheng
- Department of Ophthalmology, The Second Hospital of Jilin University, Jilin University, Changchun 130000, Jilin, China
| |
Collapse
|
4
|
Kang M, Farrell JJ, Zhu C, Park H, Kang S, Seo EH, Choi KY, Jun GR, Won S, Gim J, Lee KH, Farrer LA. Whole-genome sequencing study in Koreans identifies novel loci for Alzheimer's disease. Alzheimers Dement 2024; 20:8246-8262. [PMID: 39428694 DOI: 10.1002/alz.14128] [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] [Academic Contribution Register] [Received: 01/31/2024] [Revised: 06/06/2024] [Accepted: 06/18/2024] [Indexed: 10/22/2024]
Abstract
INTRODUCTION The genetic basis of Alzheimer's disease (AD) in Koreans is poorly understood. METHODS We performed an AD genome-wide association study using whole-genome sequence data from 3540 Koreans (1583 AD cases, 1957 controls) and single-nucleotide polymorphism array data from 2978 Japanese (1336 AD cases, 1642 controls). Significant findings were evaluated by pathway enrichment and differential gene expression analysis in brain tissue from controls and AD cases with and without dementia prior to death. RESULTS We identified genome-wide significant associations with APOE in the total sample and ROCK2 (rs76484417, p = 2.71×10-8) among APOE ε4 non-carriers. A study-wide significant association was found with aggregated rare variants in MICALL1 (MICAL like 1) (p = 9.04×10-7). Several novel AD-associated genes, including ROCK2 and MICALL1, were differentially expressed in AD cases compared to controls (p < 3.33×10-3). ROCK2 was also differentially expressed between AD cases with and without dementia (p = 1.34×10-4). DISCUSSION Our results provide insight into genetic mechanisms leading to AD and cognitive resilience in East Asians. HIGHLIGHTS Novel genome-wide significant associations for AD identified with ROCK2 and MICALL1. ROCK2 and MICALL1 are differentially expressed between AD cases and controls in the brain. This is the largest whole-genome-sequence study of AD in an East Asian population.
Collapse
Affiliation(s)
- Moonil Kang
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - John J Farrell
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Congcong Zhu
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Hyeonseul Park
- Department of Integrative Biological Sciences, Chosun University, Gwangju, Republic of Korea
| | - Sarang Kang
- Gwangju Alzheimer's and Related Dementia (GARD) Cohort Research Center, Chosun University, Dong-gu, Gwangju, Republic of Korea
| | - Eun Hyun Seo
- Gwangju Alzheimer's and Related Dementia (GARD) Cohort Research Center, Chosun University, Dong-gu, Gwangju, Republic of Korea
- Premedical Science, College of Medicine, Chosun University, Dong-gu, Gwangju, Republic of Korea
| | - Kyu Yeong Choi
- Gwangju Alzheimer's and Related Dementia (GARD) Cohort Research Center, Chosun University, Dong-gu, Gwangju, Republic of Korea
- Kolab Inc., Dong-gu, Gwangju, Republic of Korea
| | - Gyungah R Jun
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
- Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Sungho Won
- Institute of Health and Environment, Seoul National University, Gwanak-gu, Seoul, Republic of Korea
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Gwanak-gu, Seoul, Republic of Korea
- RexSoft Corps, Gwanak-gu, Seoul, Republic of Korea
| | - Jungsoo Gim
- Department of Integrative Biological Sciences, Chosun University, Gwangju, Republic of Korea
- Gwangju Alzheimer's and Related Dementia (GARD) Cohort Research Center, Chosun University, Dong-gu, Gwangju, Republic of Korea
- Department of Biomedical Science, Chosun University, Dong-gu, Gwangju, Republic of Korea
- Well-ageing Medicare Institute, Chosun University, Dong-gu, Gwangju, Republic of Korea
| | - Kun Ho Lee
- Department of Integrative Biological Sciences, Chosun University, Gwangju, Republic of Korea
- Gwangju Alzheimer's and Related Dementia (GARD) Cohort Research Center, Chosun University, Dong-gu, Gwangju, Republic of Korea
- Department of Biomedical Science, Chosun University, Dong-gu, Gwangju, Republic of Korea
- Korea Brain Research Institute, Dong-gu, Daegu, Republic of Korea
| | - Lindsay A Farrer
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Department of Ophthalmology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
- Alzheimer's Disease Research Center, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- Department of Epidemiology, Boston University School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Ala-Kokko N, Baek I, Song Y. Development of Tissue-Engineered Model of Fibrotic Scarring after Spinal Cord Injury to Study Astrocyte Activation and Neurite Outgrowth In Vitro. ACS Biomater Sci Eng 2024; 10:6545-6557. [PMID: 39259933 PMCID: PMC11480936 DOI: 10.1021/acsbiomaterials.4c01100] [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] [Academic Contribution Register] [Received: 06/16/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/13/2024]
Abstract
Traumatic spinal cord injuries (SCI) are debilitating injuries affecting twenty-seven million people worldwide and cause functional impairments. Despite decades of research and medical advancements, current treatment options for SCI remain limited, in part due to the complex pathophysiology of spinal cord lesions including cellular transformation and extracellular matrix (ECM) remodeling. Recent studies have increased focus on fibrotic scarring after SCI, and yet much remains unclear about the impact of fibrotic scarring on SCI lesion progression. Here, using collagen and decellularized spinal cord-based composite hydrogels, a three-dimensional (3D) cell culture model mimicking the fibrous core of spinal cord lesions was implemented to investigate its influence on the surrounding astrocytes. To mimic the fibrotic milieu, collagen fibril thickness was tuned using previously established temperature-controlled casting methods. In our platforms, astrocytes in fibro-mimetic hydrogels exhibited increased levels of activation markers such as glial fibrillary acidic protein and N-cadherin. Furthermore, astrocytes in fibro-mimetic hydrogels deposited more fibronectin and laminin, further hinting that astrocytes may also contribute to fibrotic scarring. These markers were decreased when Rho-ROCK and integrin β1 were inhibited via pharmacological inhibitors. Mechanistic analysis of Yes-associated protein reveals that blocking integrin β1 prevents mechanosensing of astrocytes, contributing to altered phenotypes in variable culture conditions. In the presence of these inhibitors, astrocytes increased the secretion of brain-derived neurotrophic factor, and a greater degree of dorsal root ganglia neurite infiltration into the underlying hydrogels was observed. Altogether, this study presents a novel tissue-engineered platform to study fibrotic scarring after SCI and may be a useful platform to advance our understanding of SCI lesion aggravation.
Collapse
Affiliation(s)
- Nikolas Ala-Kokko
- Department of Biomedical
Engineering, University of Arkansas, Fayetteville, Arkansas 72701, United States
| | - Inha Baek
- Department of Biomedical
Engineering, University of Arkansas, Fayetteville, Arkansas 72701, United States
| | - Younghye Song
- Department of Biomedical
Engineering, University of Arkansas, Fayetteville, Arkansas 72701, United States
| |
Collapse
|
6
|
Hassan OI, Takamiya S, Asgarihafshejani A, Fehlings MG. Bridging the gap: a translational perspective in spinal cord injury. Exp Biol Med (Maywood) 2024; 249:10266. [PMID: 39391076 PMCID: PMC11464315 DOI: 10.3389/ebm.2024.10266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/29/2024] [Accepted: 08/27/2024] [Indexed: 10/12/2024] Open
Abstract
Traumatic spinal cord injury (SCI) is a devastating and complex condition to treat with no curative options. In the past few decades, rapid advancements in our understanding of SCI pathophysiology as well as the mergence of new treatments has created more optimism. Focusing on clinical translation, this paper provides a comprehensive overview of SCI through its epidemiology, pathophysiology, currently employed management strategies, and emerging therapeutic approaches. Additionally, it emphasizes the importance of addressing the heavy quality of life (QoL) challenges faced by SCI patients and their desires, providing a basis to tailor patient-centric forms of care. Furthermore, this paper discusses the frequently encountered barriers in translation from preclinical models to clinical settings. It also seeks to summarize significant completed and ongoing SCI clinical trials focused on neuroprotective and neuroregenerative strategies. While developing a cohesive regenerative treatment strategy remains challenging, even modest improvements in sensory and motor function can offer meaningful benefits and motivation for patients coping with this highly debilitating condition.
Collapse
Affiliation(s)
- Omar Imad Hassan
- Division of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Soichiro Takamiya
- Division of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Azam Asgarihafshejani
- Division of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
| | - Michael G. Fehlings
- Division of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
- Division of Neurosurgery and Spine Program, Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
7
|
Doody NE, Smith NJ, Akam EC, Askew GN, Kwok JCF, Ichiyama RM. Differential expression of genes in the RhoA/ROCK pathway in the hippocampus and cortex following intermittent hypoxia and high-intensity interval training. J Neurophysiol 2024; 132:531-543. [PMID: 38985935 PMCID: PMC11427053 DOI: 10.1152/jn.00422.2023] [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] [Academic Contribution Register] [Received: 11/14/2023] [Revised: 06/13/2024] [Accepted: 07/04/2024] [Indexed: 07/12/2024] Open
Abstract
Structural neuroplasticity such as neurite extension and dendritic spine dynamics is enhanced by brain-derived neurotrophic factor (BDNF) and impaired by types of inhibitory molecules that induce growth cone collapse and actin depolymerization, for example, myelin-associated inhibitors, chondroitin sulfate proteoglycans, and negative guidance molecules. These inhibitory molecules can activate RhoA/rho-associated coiled-coil containing protein kinase (ROCK) signaling (known to restrict structural plasticity). Intermittent hypoxia (IH) and high-intensity interval training (HIIT) are known to upregulate BDNF that is associated with improvements in learning and memory and greater functional recovery following neural insults. We investigated whether the RhoA/ROCK signaling pathway is also modulated by IH and HIIT in the hippocampus, cortex, and lumbar spinal cord of male Wistar rats. The gene expression of 25 RhoA/ROCK signaling pathway components was determined following IH, HIIT, or IH combined with HIIT (30 min/day, 5 days/wk, 6 wk). IH included 10 3-min bouts that alternated between hypoxia (15% O2) and normoxia. HIIT included 10 3-min bouts alternating between treadmill speeds of 50 cm·s-1 and 15 cm·s-1. In the hippocampus, IH and HIIT significantly downregulated Acan and NgR2 mRNA that are involved in the inhibition of neuroplasticity. However, IH and IH + HIIT significantly upregulated Lingo-1 and NgR3 in the cortex. This is the first time IH and HIIT have been linked to the modulation of plasticity-inhibiting pathways. These results provide a fundamental step toward elucidating the interplay between the neurotrophic and inhibitory mechanisms involved in experience-driven neural plasticity that will aid in optimizing physiological interventions for the treatment of cognitive decline or neurorehabilitation.NEW & NOTEWORTHY Intermittent hypoxia (IH) and high-intensity interval training (HIIT) enhance neuroplasticity and upregulate neurotrophic factors in the central nervous system (CNS). We provide evidence that IH and IH + HIIT also have the capacity to regulate genes involved in the RhoA/ROCK signaling pathway that is known to restrict structural plasticity in the CNS. This provides a new mechanistic insight into how these interventions may enhance hippocampal-related plasticity and facilitate learning, memory, and neuroregeneration.
Collapse
Affiliation(s)
- Natalie E Doody
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, United Kingdom
| | - Nicole J Smith
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
- Department of Cell and Molecular Biology, Karolinska Institute, Stockholm, Sweden
| | - Elizabeth C Akam
- School of Sport, Exercise, and Health Sciences, Loughborough University, Loughborough, United Kingdom
| | - Graham N Askew
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Jessica C F Kwok
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
- Institute of Experimental Medicine, The Czech Academy of Sciences, Prague 4, Czech Republic
| | - Ronaldo M Ichiyama
- School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
8
|
Alvi MA, Pedro KM, Quddusi AI, Fehlings MG. Advances and Challenges in Spinal Cord Injury Treatments. J Clin Med 2024; 13:4101. [PMID: 39064141 PMCID: PMC11278467 DOI: 10.3390/jcm13144101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/16/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Spinal cord injury (SCI) is a debilitating condition that is associated with long-term physical and functional disability. Our understanding of the pathogenesis of SCI has evolved significantly over the past three decades. In parallel, significant advances have been made in optimizing the management of patients with SCI. Early surgical decompression, adequate bony decompression and expansile duraplasty are surgical strategies that may improve neurological and functional outcomes in patients with SCI. Furthermore, advances in the non-surgical management of SCI have been made, including optimization of hemodynamic management in the critical care setting. Several promising therapies have also been investigated in pre-clinical studies, with some being translated into clinical trials. Given the recent interest in advancing precision medicine, several investigations have been performed to delineate the role of imaging, cerebral spinal fluid (CSF) and serum biomarkers in predicting outcomes and curating individualized treatment plans for SCI patients. Finally, technological advancements in biomechanics and bioengineering have also found a role in SCI management in the form of neuromodulation and brain-computer interfaces.
Collapse
Affiliation(s)
- Mohammed Ali Alvi
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; (M.A.A.); (K.M.P.); (A.I.Q.)
| | - Karlo M. Pedro
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; (M.A.A.); (K.M.P.); (A.I.Q.)
- Department of Surgery and Spine Program, University of Toronto, Toronto, ON M5T 1P5, Canada
| | - Ayesha I. Quddusi
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; (M.A.A.); (K.M.P.); (A.I.Q.)
| | - Michael G. Fehlings
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada; (M.A.A.); (K.M.P.); (A.I.Q.)
- Department of Surgery and Spine Program, University of Toronto, Toronto, ON M5T 1P5, Canada
- Division of Neurosurgery, Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network, Toronto, ON M5T 2S8, Canada
| |
Collapse
|
9
|
Sun Y, Xiao Z, Chen B, Zhao Y, Dai J. Advances in Material-Assisted Electromagnetic Neural Stimulation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2400346. [PMID: 38594598 DOI: 10.1002/adma.202400346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 01/08/2024] [Revised: 03/26/2024] [Indexed: 04/11/2024]
Abstract
Bioelectricity plays a crucial role in organisms, being closely connected to neural activity and physiological processes. Disruptions in the nervous system can lead to chaotic ionic currents at the injured site, causing disturbances in the local cellular microenvironment, impairing biological pathways, and resulting in a loss of neural functions. Electromagnetic stimulation has the ability to generate internal currents, which can be utilized to counter tissue damage and aid in the restoration of movement in paralyzed limbs. By incorporating implanted materials, electromagnetic stimulation can be targeted more accurately, thereby significantly improving the effectiveness and safety of such interventions. Currently, there have been significant advancements in the development of numerous promising electromagnetic stimulation strategies with diverse materials. This review provides a comprehensive summary of the fundamental theories, neural stimulation modulating materials, material application strategies, and pre-clinical therapeutic effects associated with electromagnetic stimulation for neural repair. It offers a thorough analysis of current techniques that employ materials to enhance electromagnetic stimulation, as well as potential therapeutic strategies for future applications.
Collapse
Affiliation(s)
- Yuting Sun
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300192, China
| |
Collapse
|
10
|
Kvistad CE, Kråkenes T, Gavasso S, Bø L. Neural regeneration in the human central nervous system-from understanding the underlying mechanisms to developing treatments. Where do we stand today? Front Neurol 2024; 15:1398089. [PMID: 38803647 PMCID: PMC11129638 DOI: 10.3389/fneur.2024.1398089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/08/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024] Open
Abstract
Mature neurons in the human central nervous system (CNS) fail to regenerate after injuries. This is a common denominator across different aetiologies, including multiple sclerosis, spinal cord injury and ischemic stroke. The lack of regeneration leads to permanent functional deficits with a substantial impact on patient quality of life, representing a significant socioeconomic burden worldwide. Great efforts have been made to decipher the responsible mechanisms and we now know that potent intra- and extracellular barriers prevent axonal repair. This knowledge has resulted in numerous clinical trials, aiming to promote neuroregeneration through different approaches. Here, we summarize the current understanding of the causes to the poor regeneration within the human CNS. We also review the results of the treatment attempts that have been translated into clinical trials so far.
Collapse
Affiliation(s)
| | - Torbjørn Kråkenes
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Sonia Gavasso
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Lars Bø
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, Bergen, Norway
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
11
|
Collu R, Yin Z, Giunti E, Daley S, Chen M, Morin P, Killick R, Wong STC, Xia W. Effect of the ROCK inhibitor fasudil on the brain proteomic profile in the tau transgenic mouse model of Alzheimer's disease. Front Aging Neurosci 2024; 16:1323563. [PMID: 38440100 PMCID: PMC10911083 DOI: 10.3389/fnagi.2024.1323563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/18/2023] [Accepted: 01/29/2024] [Indexed: 03/06/2024] Open
Abstract
Introduction The goal of this study is to explore the pharmacological potential of the amyloid-reducing vasodilator fasudil, a selective Ras homolog (Rho)-associated kinases (ROCK) inhibitor, in the P301S tau transgenic mouse model (Line PS19) of neurodegenerative tauopathy and Alzheimer's disease (AD). Methods We used LC-MS/MS, ELISA and bioinformatic approaches to investigate the effect of treatment with fasudil on the brain proteomic profile in PS19 tau transgenic mice. We also explored the efficacy of fasudil in reducing tau phosphorylation, and the potential beneficial and/or toxic effects of its administration in mice. Results Proteomic profiling of mice brains exposed to fasudil revealed the activation of the mitochondrial tricarboxylic acid (TCA) cycle and blood-brain barrier (BBB) gap junction metabolic pathways. We also observed a significant negative correlation between the brain levels of phosphorylated tau (pTau) at residue 396 and both fasudil and its metabolite hydroxyfasudil. Conclusions Our results provide evidence on the activation of proteins and pathways related to mitochondria and BBB functions by fasudil treatment and support its further development and therapeutic potential for AD.
Collapse
Affiliation(s)
- Roberto Collu
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, United States
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Zheng Yin
- T. T. and W. F. Chao Center for BRAIN, Houston Methodist Hospital, Houston Methodist Academic Institute, Houston, TX, United States
| | - Elisa Giunti
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, United States
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Sarah Daley
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, United States
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Mei Chen
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, United States
| | - Peter Morin
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
| | - Richard Killick
- King's College London, Maurice Wohl Clinical Neuroscience Institute, London, United Kingdom
| | - Stephen T. C. Wong
- T. T. and W. F. Chao Center for BRAIN, Houston Methodist Hospital, Houston Methodist Academic Institute, Houston, TX, United States
| | - Weiming Xia
- Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, United States
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian and Avedisian School of Medicine, Boston, MA, United States
- Department of Biological Sciences, University of Massachusetts Kennedy College of Science, Lowell, MA, United States
| |
Collapse
|
12
|
Cai Z, Zhang Y, Fang RS, Brenner B, Kweon J, Sun C, Goldberg JL, Zhang HF. Multiscale imaging of corneal endothelium damage and Rho-kinase inhibitor application in mouse models of acute ocular hypertension. BIOMEDICAL OPTICS EXPRESS 2024; 15:1102-1114. [PMID: 38404323 PMCID: PMC10890882 DOI: 10.1364/boe.510432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Academic Contribution Register] [Received: 10/26/2023] [Revised: 01/13/2024] [Accepted: 01/15/2024] [Indexed: 02/27/2024]
Abstract
We developed a multiscale optical imaging workflow, integrating and correlating visible-light optical coherence tomography, confocal laser scanning microscopy, and single-molecule localization microscopy to investigate mouse cornea damage from the in-vivo tissue level to the nanoscopic single-molecule level. We used electron microscopy to validate the imaged nanoscopic structures. We imaged wild-type mice and mice with acute ocular hypertension and examined the effects of Rho-kinase inhibitor application. We defined four types of intercellular tight junction structures as healthy, compact, partially-distorted, and fully-distorted types by labeling the zonula occludens-1 protein in the corneal endothelial cell layer. We correlated the statistics of the four types of tight junction structures with cornea thickness and intraocular pressure. We found that the population of fully-distorted tight junctions correlated well with the level of corneal edema, and applying Rho-kinase inhibitor reduced the population of fully-distorted tight junctions under acute ocular hypertension. Together, these data point to the utility of multiscale optical imaging in revealing fundamental biology relevant to disease and therapeutics.
Collapse
Affiliation(s)
- Zhen Cai
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Currently with Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Zhang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
- Currently with Program of Polymer and Color Chemistry, Department of Textile Engineering, Chemistry and Science, North Carolina State University, Raleigh, NC 27606, USA
| | - Raymond S. Fang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Benjamin Brenner
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Junghun Kweon
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Cheng Sun
- Department of Mechanical Engineering, Northwestern University, Evanston, IL 60208, USA
| | - Jeffrey L. Goldberg
- Spencer Center for Vision Research, Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, CA 94303, USA
| | - Hao F. Zhang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, USA
| |
Collapse
|
13
|
Maroto IB, Moreno E, Costas-Insua C, Merino-Gracia J, Diez-Alarcia R, Álvaro-Blázquez A, Canales Á, Canela EI, Casadó V, Urigüen L, Rodríguez-Crespo I, Guzmán M. Selective inhibition of cannabinoid CB 1 receptor-evoked signalling by the interacting protein GAP43. Neuropharmacology 2023; 240:109712. [PMID: 37689260 DOI: 10.1016/j.neuropharm.2023.109712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/24/2023] [Revised: 09/05/2023] [Accepted: 09/06/2023] [Indexed: 09/11/2023]
Abstract
Cannabinoids exert pleiotropic effects on the brain by engaging the cannabinoid CB1 receptor (CB1R), a presynaptic metabotropic receptor that regulates key neuronal functions in a highly context-dependent manner. We have previously shown that CB1R interacts with growth-associated protein of 43 kDa (GAP43) and that this interaction inhibits CB1R function on hippocampal excitatory synaptic transmission, thereby impairing the therapeutic effect of cannabinoids on epileptic seizures in vivo. However, the underlying molecular features of this interaction remain unexplored. Here, we conducted mechanistic experiments on HEK293T cells co-expressing CB1R and GAP43 and show that GAP43 modulates CB1R signalling in a strikingly selective manner. Specifically, GAP43 did not affect the archetypical agonist-evoked (i) CB1R/Gi/o protein-coupled signalling pathways, such as cAMP/PKA and ERK, or (ii) CB1R internalization and intracellular trafficking. In contrast, GAP43 blocked an alternative agonist-evoked CB1R-mediated activation of the cytoskeleton-associated ROCK signalling pathway, which relied on the GAP43-mediated impairment of CB1R/Gq/11 protein coupling. GAP43 also abrogated CB1R-mediated ROCK activation in mouse hippocampal neurons, and this process led in turn to a blockade of cannabinoid-evoked neurite collapse. An NMR-based characterization of the CB1R-GAP43 interaction supported that GAP43 binds directly and specifically through multiple amino acid stretches to the C-terminal domain of the receptor. Taken together, our findings unveil a CB1R-Gq/11-ROCK signalling axis that is selectively impaired by GAP43 and may ultimately control neurite outgrowth.
Collapse
Affiliation(s)
- Irene B Maroto
- Department of Biochemistry and Molecular Biology, Instituto Universitario de Investigación Neuroquímica (IUIN), Complutense University, 28040, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034, Madrid, Spain
| | - Estefanía Moreno
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology and Institute of Biomedicine of the University of Barcelona, University of Barcelona, 08028, Barcelona, Spain
| | - Carlos Costas-Insua
- Department of Biochemistry and Molecular Biology, Instituto Universitario de Investigación Neuroquímica (IUIN), Complutense University, 28040, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034, Madrid, Spain
| | - Javier Merino-Gracia
- Department of Biochemistry and Molecular Biology, Instituto Universitario de Investigación Neuroquímica (IUIN), Complutense University, 28040, Madrid, Spain
| | - Rebeca Diez-Alarcia
- Department of Pharmacology, University of the Basque Country/Euskal Herriko Unibertsitatea, 48940, Leioa, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), 28029, Madrid, Spain; Biocruces Bizkaia Health Research Institute, 48903, Barakaldo, Bizkaia, Spain
| | - Alicia Álvaro-Blázquez
- Department of Biochemistry and Molecular Biology, Instituto Universitario de Investigación Neuroquímica (IUIN), Complutense University, 28040, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034, Madrid, Spain
| | - Ángeles Canales
- Department of Organic Chemistry, Instituto Universitario de Investigación Neuroquímica (IUIN), Complutense University, 28040, Madrid, Spain
| | - Enric I Canela
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology and Institute of Biomedicine of the University of Barcelona, University of Barcelona, 08028, Barcelona, Spain
| | - Vicent Casadó
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology and Institute of Biomedicine of the University of Barcelona, University of Barcelona, 08028, Barcelona, Spain
| | - Leyre Urigüen
- Department of Pharmacology, University of the Basque Country/Euskal Herriko Unibertsitatea, 48940, Leioa, Spain; Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), 28029, Madrid, Spain; Biocruces Bizkaia Health Research Institute, 48903, Barakaldo, Bizkaia, Spain
| | - Ignacio Rodríguez-Crespo
- Department of Biochemistry and Molecular Biology, Instituto Universitario de Investigación Neuroquímica (IUIN), Complutense University, 28040, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034, Madrid, Spain
| | - Manuel Guzmán
- Department of Biochemistry and Molecular Biology, Instituto Universitario de Investigación Neuroquímica (IUIN), Complutense University, 28040, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, 28029, Madrid, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28034, Madrid, Spain.
| |
Collapse
|
14
|
Mannan A, Dhiamn S, Garg N, Singh TG. Pharmacological modulation of Sonic Hedgehog signaling pathways in Angiogenesis: A mechanistic perspective. Dev Biol 2023; 504:58-74. [PMID: 37739118 DOI: 10.1016/j.ydbio.2023.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/01/2023] [Revised: 09/13/2023] [Accepted: 09/18/2023] [Indexed: 09/24/2023]
Abstract
The Sonic hedgehog (SHh) signaling pathway is an imperative operating network that helps in regulates the critical events during the development processes like multicellular embryo growth and patterning. Disruptions in SHh pathway regulation can have severe consequences, including congenital disabilities, stem cell renewal, tissue regeneration, and cancer/tumor growth. Activation of the SHh signal occurs when SHh binds to the receptor complex of Patch (Ptc)-mediated Smoothened (Smo) (Ptc-smo), initiating downstream signaling. This review explores how pharmacological modulation of the SHh pathway affects angiogenesis through canonical and non-canonical pathways. The canonical pathway for angiogenesis involves the activation of angiogenic cytokines such as fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), placental growth factor (PGF), hepatocyte growth factor (HGF), platelet-derived growth factor (PDGF), stromal cell-derived factor 1α, transforming growth factor-β1 (TGF-β1), and angiopoietins (Ang-1 and Ang-2), which facilitate the process of angiogenesis. The Non-canonical pathway includes indirect activation of certain pathways like iNOS/Netrin-1/PKC, RhoA/Rock, ERK/MAPK, PI3K/Akt, Wnt/β-catenin, Notch signaling pathway, and so on. This review will provide a better grasp of the mechanistic approach of SHh in mediating angiogenesis, which can aid in the suppression of certain cancer and tumor growths.
Collapse
Affiliation(s)
- Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Sonia Dhiamn
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Nikhil Garg
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, 140401, Punjab, India.
| |
Collapse
|
15
|
Corallo D, Dalla Vecchia M, Lazic D, Taschner-Mandl S, Biffi A, Aveic S. The molecular basis of tumor metastasis and current approaches to decode targeted migration-promoting events in pediatric neuroblastoma. Biochem Pharmacol 2023; 215:115696. [PMID: 37481138 DOI: 10.1016/j.bcp.2023.115696] [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] [Academic Contribution Register] [Received: 04/23/2023] [Revised: 07/12/2023] [Accepted: 07/12/2023] [Indexed: 07/24/2023]
Abstract
Cell motility is a crucial biological process that plays a critical role in the development of multicellular organisms and is essential for tissue formation and regeneration. However, uncontrolled cell motility can lead to the development of various diseases, including neoplasms. In this review, we discuss recent advances in the discovery of regulatory mechanisms underlying the metastatic spread of neuroblastoma, a solid pediatric tumor that originates in the embryonic migratory cells of the neural crest. The highly motile phenotype of metastatic neuroblastoma cells requires targeting of intracellular and extracellular processes, that, if affected, would be helpful for the treatment of high-risk patients with neuroblastoma, for whom current therapies remain inadequate. Development of new potentially migration-inhibiting compounds and standardized preclinical approaches for the selection of anti-metastatic drugs in neuroblastoma will also be discussed.
Collapse
Affiliation(s)
- Diana Corallo
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padova, Italy
| | - Marco Dalla Vecchia
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padova, Italy
| | - Daria Lazic
- St. Anna Children's Cancer Research Institute, CCRI, Zimmermannplatz 10, 1090, Vienna, Austria
| | - Sabine Taschner-Mandl
- St. Anna Children's Cancer Research Institute, CCRI, Zimmermannplatz 10, 1090, Vienna, Austria
| | - Alessandra Biffi
- Pediatric Hematology, Oncology and Stem Cell Transplant Division, Woman's and Child Health Department, University of Padova, 35121 Padova, Italy
| | - Sanja Aveic
- Laboratory of Target Discovery and Biology of Neuroblastoma, Istituto di Ricerca Pediatrica (IRP), Fondazione Città della Speranza, 35127 Padova, Italy.
| |
Collapse
|
16
|
Punjani N, Deska-Gauthier D, Hachem LD, Abramian M, Fehlings MG. Neuroplasticity and regeneration after spinal cord injury. NORTH AMERICAN SPINE SOCIETY JOURNAL 2023; 15:100235. [PMID: 37416090 PMCID: PMC10320621 DOI: 10.1016/j.xnsj.2023.100235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 04/13/2023] [Revised: 06/05/2023] [Accepted: 06/05/2023] [Indexed: 07/08/2023]
Abstract
Spinal cord injury (SCI) is a debilitating condition with significant personal, societal, and economic burden. The highest proportion of traumatic injuries occur at the cervical level, which results in severe sensorimotor and autonomic deficits. Following the initial physical damage associated with traumatic injuries, secondary pro-inflammatory, excitotoxic, and ischemic cascades are initiated further contributing to neuronal and glial cell death. Additionally, emerging evidence has begun to reveal that spinal interneurons undergo subtype specific neuroplastic circuit rearrangements in the weeks to months following SCI, contributing to or hindering functional recovery. The current therapeutic guidelines and standards of care for SCI patients include early surgery, hemodynamic regulation, and rehabilitation. Additionally, preclinical work and ongoing clinical trials have begun exploring neuroregenerative strategies utilizing endogenous neural stem/progenitor cells, stem cell transplantation, combinatorial approaches, and direct cell reprogramming. This review will focus on emerging cellular and noncellular regenerative therapies with an overview of the current available strategies, the role of interneurons in plasticity, and the exciting research avenues enhancing tissue repair following SCI.
Collapse
Affiliation(s)
- Nayaab Punjani
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Dylan Deska-Gauthier
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Laureen D. Hachem
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Surgery, Division of Neurosurgery and Spine Program, University of Toronto, Toronto, ON, Canada
| | - Madlene Abramian
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Michael G. Fehlings
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Surgery, Division of Neurosurgery and Spine Program, University of Toronto, Toronto, ON, Canada
- Division of Neurosurgery, Krembil Neuroscience Centre, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| |
Collapse
|
17
|
Sousa T, Moreira PI, Cardoso S. Current Advances in Mitochondrial Targeted Interventions in Alzheimer's Disease. Biomedicines 2023; 11:2331. [PMID: 37760774 PMCID: PMC10525414 DOI: 10.3390/biomedicines11092331] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/11/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 09/29/2023] Open
Abstract
Alzheimer's disease is the most prevalent neurodegenerative disorder and affects the lives not only of those who are diagnosed but also of their caregivers. Despite the enormous social, economic and political burden, AD remains a disease without an effective treatment and with several failed attempts to modify the disease course. The fact that AD clinical diagnosis is most often performed at a stage at which the underlying pathological events are in an advanced and conceivably irremediable state strongly hampers treatment attempts. This raises the awareness of the need to identify and characterize the early brain changes in AD, in order to identify possible novel therapeutic targets to circumvent AD's cascade of events. One of the most auspicious targets is mitochondria, powerful organelles found in nearly all cells of the body. A vast body of literature has shown that mitochondria from AD patients and model organisms of the disease differ from their non-AD counterparts. In view of this evidence, preserving and/or restoring mitochondria's health and function can represent the primary means to achieve advances to tackle AD. In this review, we will briefly assess and summarize the previous and latest evidence of mitochondria dysfunction in AD. A particular focus will be given to the recent updates and advances in the strategy options aimed to target faulty mitochondria in AD.
Collapse
Affiliation(s)
- Tiago Sousa
- Faculty of Medicine, University of Coimbra, 3000-370 Coimbra, Portugal;
| | - Paula I. Moreira
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-370 Coimbra, Portugal
| | - Susana Cardoso
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal;
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- IIIUC—Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal
| |
Collapse
|
18
|
Mohan M, Mannan A, Singh TG. Therapeutic implication of Sonic Hedgehog as a potential modulator in ischemic injury. Pharmacol Rep 2023:10.1007/s43440-023-00505-0. [PMID: 37347388 DOI: 10.1007/s43440-023-00505-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/21/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/23/2023]
Abstract
Sonic Hedgehog (SHh) is a homology protein that is involved in the modeling and development of embryonic tissues. As SHh plays both protective and harmful roles in ischemia, any disruption in the transduction and regulation of the SHh signaling pathway causes ischemia to worsen. The SHh signal activation occurs when SHh binds to the receptor complex of Ptc-mediated Smoothened (Smo) (Ptc-smo), which initiates the downstream signaling cascade. This article will shed light on how pharmacological modifications to the SHh signaling pathway transduction mechanism alter ischemic conditions via canonical and non-canonical pathways by activating certain downstream signaling cascades with respect to protein kinase pathways, angiogenic cytokines, inflammatory mediators, oxidative parameters, and apoptotic pathways. The canonical pathway includes direct activation of interleukins (ILs), angiogenic cytokines like hepatocyte growth factor (HGF), platelet-derived growth factor (PDGF), vascular endothelial growth factor (VEGF), epidermal growth factor (EGF), and hypoxia-inducible factor alpha (HIF-), which modulate ischemia. The non-canonical pathway includes indirect activation of certain pathways like mTOR, PI3K/Akt, MAPK, RhoA/ROCK, Wnt/-catenin, NOTCH, Forkhead box protein (FOXF), Toll-like receptors (TLR), oxidative parameters such as GSH, SOD, and CAT, and some apoptotic parameters such as Bcl2. This review provides comprehensive insights that contribute to our knowledge of how SHh impacts the progression and outcomes of ischemic injuries.
Collapse
Affiliation(s)
- Maneesh Mohan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
19
|
Kim HJ, Hwang JS, Noh KB, Oh SH, Park JB, Shin YJ. A p-Tyr42 RhoA Inhibitor Promotes the Regeneration of Human Corneal Endothelial Cells by Ameliorating Cellular Senescence. Antioxidants (Basel) 2023; 12:1186. [PMID: 37371916 DOI: 10.3390/antiox12061186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/14/2023] [Revised: 05/18/2023] [Accepted: 05/27/2023] [Indexed: 06/29/2023] Open
Abstract
The development of treatment strategies for human corneal endothelial cells (hCECs) disease is necessary because hCECs do not regenerate in vivo due to the properties that are similar to senescence. This study is performed to investigate the role of a p-Tyr42 RhoA inhibitor (MH4, ELMED Inc., Chuncheon) in transforming growth factor-beta (TGF-β)- or H2O2-induced cellular senescence of hCECs. Cultured hCECs were treated with MH4. The cell shape, proliferation rate, and cell cycle phases were analyzed. Moreover, cell adhesion assays and immunofluorescence staining for F-actin, Ki-67, and E-cadherin were performed. Additionally, the cells were treated with TGF-β or H2O2 to induce senescence, and mitochondrial oxidative reactive oxygen species (ROS) levels, mitochondrial membrane potential, and NF-κB translocation were evaluated. LC3II/LC3I levels were determined using Western blotting to analyze autophagy. MH4 promotes hCEC proliferation, shifts the cell cycle, attenuates actin distribution, and increases E-cadherin expression. TGF-β and H2O2 induce senescence by increasing mitochondrial ROS levels and NF-κB translocation into the nucleus; however, this effect is attenuated by MH4. Moreover, TGF-β and H2O2 decrease the mitochondrial membrane potential and induce autophagy, while MH4 reverses these effects. In conclusion, MH4, a p-Tyr42 RhoA inhibitor, promotes the regeneration of hCECs and protects hCECs against TGF-β- and H2O2-induced senescence via the ROS/NF-κB/mitochondrial pathway.
Collapse
Affiliation(s)
- Hyeon Jung Kim
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07442, Republic of Korea
- Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul 07442, Republic of Korea
| | - Jin Sun Hwang
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07442, Republic of Korea
- Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul 07442, Republic of Korea
| | - Kyung Bo Noh
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07442, Republic of Korea
- Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul 07442, Republic of Korea
| | - Sun-Hee Oh
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07442, Republic of Korea
- Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul 07442, Republic of Korea
| | - Jae-Bong Park
- Department of Biochemistry, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea
| | - Young Joo Shin
- Department of Ophthalmology, Hallym University Medical Center, Hallym University College of Medicine, Seoul 07442, Republic of Korea
- Hallym BioEyeTech Research Center, Hallym University College of Medicine, Seoul 07442, Republic of Korea
| |
Collapse
|
20
|
Cai Z, Zhang Y, Fang RS, Brenner B, Kweon J, Sun C, Goldberg J, Zhang HF. Multiscale imaging of corneal endothelium damage and effects of Rho Kinase inhibitor application in mouse models of acute ocular hypertension. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.18.541299. [PMID: 37292938 PMCID: PMC10245768 DOI: 10.1101/2023.05.18.541299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 06/10/2023]
Abstract
We developed a multiscale optical imaging workflow, integrating and correlating visible-light optical coherence tomography, confocal laser scanning microscopy, and single-molecule localization microscopy to investigate the mouse cornea damages from the in-vivo tissue level to the nanoscopic single-molecule level. We used electron microscopy to validate the imaged nanoscopic structures. We imaged wild-type mice and mice with acute ocular hypertension and examined the effects of Rho Kinase inhibitor application. We defined four types of intercellular tight junction structures as healthy, compact, partially-distorted, and fully-distorted types by labeling the Zonula occludens-1 protein in the corneal endothelial cell layer. We correlated the statistics of the four types of tight junction structures with cornea thickness and intraocular pressure. We found that the population of fully-distorted tight junctions correlated well with the level of cornea edema, and applying Rho Kinase inhibitor reduced the population of fully-distorted tight junctions under acute ocular hypertension.
Collapse
|
21
|
Mani S, Jindal D, Chopra H, Jha SK, Singh SK, Ashraf GM, Kamal M, Iqbal D, Chellappan DK, Dey A, Dewanjee S, Singh KK, Ojha S, Singh I, Gautam RK, Jha NK. ROCK2 Inhibition: A Futuristic Approach for the Management of Alzheimer's Disease. Neurosci Biobehav Rev 2022; 142:104871. [PMID: 36122738 DOI: 10.1016/j.neubiorev.2022.104871] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/12/2022] [Revised: 07/30/2022] [Accepted: 09/12/2022] [Indexed: 12/06/2022]
Abstract
Neurons depend on mitochondrial functions for membrane excitability, neurotransmission, and plasticity.Mitochondrialdynamicsare important for neural cell maintenance. To maintain mitochondrial homeostasis, lysosomes remove dysfunctionalmitochondria through mitophagy. Mitophagy promotes mitochondrial turnover and prevents the accumulation of dysfunctional mitochondria. In many neurodegenerative diseases (NDDs), including Alzheimer's disease (AD), mitophagy is disrupted in neurons.Mitophagy is regulated by several proteins; recently,Rho-associated coiled-coil containing protein kinase 2 (ROCK2) has been suggested to negatively regulate the Parkin-dependent mitophagy pathway.Thus, ROCK2inhibitionmay bea promising therapyfor NDDs. This review summarizesthe mitophagy pathway, the role of ROCK2in Parkin-dependentmitophagyregulation,and mitophagy impairment in the pathology of AD. We further discuss different ROCK inhibitors (synthetic drugs, natural compounds,and genetherapy-based approaches)and examine their effects on triggering neuronal growth and neuroprotection in AD and other NDDs. This comprehensive overview of the role of ROCK in mitophagy inhibition provides a possible explanation for the significance of ROCK inhibitors in the therapeutic management of AD and other NDDs.
Collapse
Affiliation(s)
- Shalini Mani
- Centre for Emerging Disease, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India.
| | - Divya Jindal
- Centre for Emerging Disease, Department of Biotechnology, Jaypee Institute of Information Technology, Noida, UP, India
| | - Hitesh Chopra
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, Uttar Pradesh 201310, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, 140413, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, Punjab, India
| | | | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Danish Iqbal
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Abhijit Dey
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, India
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Keshav K Singh
- Department of Genetics, UAB School of Medicine, The University of Alabama at Birmingham
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, United Arab Emirates
| | - Inderbir Singh
- MM School of Pharmacy, MM University, Sadopur-Ambala -134007, India
| | - Rupesh K Gautam
- MM School of Pharmacy, MM University, Sadopur-Ambala -134007, India.
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida, Uttar Pradesh 201310, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, 140413, India.
| |
Collapse
|
22
|
The Relationship between Cadherin Polymorphisms and the Risk of Delayed Encephalopathy after Acute Carbon Monoxide Poisoning in the Chinese Han Population. Behav Neurol 2022; 2022:3155703. [PMID: 35140817 PMCID: PMC8818434 DOI: 10.1155/2022/3155703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/16/2021] [Accepted: 01/15/2022] [Indexed: 11/17/2022] Open
Abstract
Objective The purpose of this study was to analyze the relationship between cadherin gene single-nucleotide polymorphisms (SNPs) and the risk of delayed encephalopathy after acute carbon monoxide poisoning (DEACMP). Materials and Methods A total of 416 patients with DEACMP and 754 patients with acute carbon monoxide poisoning (ACMP) were recruited. We used the Sequenom MassARRAY® system to detect cadherin gene SNPs related to DEACMP. Using different genetic analysis models, we evaluated the relationship between the cadherin gene polymorphisms and risk of DEACMP. Results We found that rs1944294 in the N-cadherin (CDH2) gene showed significant differences in genotype frequencies between the two groups under codominant and dominant inheritance models. Similarly, rs2513796 in the cadherin-17 (CDH17) gene showed significant differences under the codominant, dominant, and overdominant genetic models. And the T allele frequency of rs1944294 in the DEACMP group was significantly higher than that in the ACMP group (P = 0.023). Conclusions Cadherin gene SNPs (rs1944294, rs2513796) are associated with an increased risk of DEACMP in the Chinese population.
Collapse
|
23
|
Miller B, Sewell-Loftin MK. Mechanoregulation of Vascular Endothelial Growth Factor Receptor 2 in Angiogenesis. Front Cardiovasc Med 2022; 8:804934. [PMID: 35087885 PMCID: PMC8787114 DOI: 10.3389/fcvm.2021.804934] [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] [Subscribe] [Academic Contribution Register] [Received: 10/29/2021] [Accepted: 12/10/2021] [Indexed: 12/17/2022] Open
Abstract
The endothelial cells that compose the vascular system in the body display a wide range of mechanotransductive behaviors and responses to biomechanical stimuli, which act in concert to control overall blood vessel structure and function. Such mechanosensitive activities allow blood vessels to constrict, dilate, grow, or remodel as needed during development as well as normal physiological functions, and the same processes can be dysregulated in various disease states. Mechanotransduction represents cellular responses to mechanical forces, translating such factors into chemical or electrical signals which alter the activation of various cell signaling pathways. Understanding how biomechanical forces drive vascular growth in healthy and diseased tissues could create new therapeutic strategies that would either enhance or halt these processes to assist with treatments of different diseases. In the cardiovascular system, new blood vessel formation from preexisting vasculature, in a process known as angiogenesis, is driven by vascular endothelial growth factor (VEGF) binding to VEGF receptor 2 (VEGFR-2) which promotes blood vessel development. However, physical forces such as shear stress, matrix stiffness, and interstitial flow are also major drivers and effectors of angiogenesis, and new research suggests that mechanical forces may regulate VEGFR-2 phosphorylation. In fact, VEGFR-2 activation has been linked to known mechanobiological agents including ERK/MAPK, c-Src, Rho/ROCK, and YAP/TAZ. In vascular disease states, endothelial cells can be subjected to altered mechanical stimuli which affect the pathways that control angiogenesis. Both normalizing and arresting angiogenesis associated with tumor growth have been strategies for anti-cancer treatments. In the field of regenerative medicine, harnessing biomechanical regulation of angiogenesis could enhance vascularization strategies for treating a variety of cardiovascular diseases, including ischemia or permit development of novel tissue engineering scaffolds. This review will focus on the impact of VEGFR-2 mechanosignaling in endothelial cells (ECs) and its interaction with other mechanotransductive pathways, as well as presenting a discussion on the relationship between VEGFR-2 activation and biomechanical forces in the extracellular matrix (ECM) that can help treat diseases with dysfunctional vascular growth.
Collapse
Affiliation(s)
- Bronte Miller
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mary Kathryn Sewell-Loftin
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States.,O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
24
|
Wang J, Zheng B, Yang S, Zheng H, Wang J. Opicapone Protects Against Hyperhomocysteinemia-Induced Increase in Blood-Brain Barrier Permeability. Neurotox Res 2021; 39:2018-2028. [PMID: 34709593 DOI: 10.1007/s12640-021-00429-8] [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] [Academic Contribution Register] [Received: 07/21/2021] [Revised: 09/28/2021] [Accepted: 10/06/2021] [Indexed: 10/20/2022]
Abstract
Hyperhomocysteinemia (HHcy)-related brain vascular disorders and brain endothelial dysfunction are important characteristics of the pathogeneses of subarachnoid hemorrhage and stroke. Upregulated homocysteine (Hcy) can impair the integrity of the blood-brain barrier (BBB). Opicapone has been recently licensed for the management of Parkinson's disease (PD); however, it is unknown whether it possesses a protective effect in brain vessels against HHcy. To investigate the beneficial effects of Opicapone on BBB permeability against HHcy, we carried out both in vivo and in vitro experiments. Mice were allocated into four groups: the Control, Opicapone, HHcy, and HHcy + Opicapone. Interestingly, we found that the administration of Opicapone attenuated the increased BBB permeability in Hcy-treated mice, as determined by sodium fluorescein staining. The immunofluorescence staining showed that Opicapone prevented homocysteine-induced reduction of claudin-2 in the mice cortices. The in situ zymography assay revealed that Opicapone suppressed homocysteine-increased matrix metalloproteinases (MMPs) activity in the cortices. In bEnd.3 brain endothelial cells, Opicapone treatment ameliorated homocysteine-induced lactate dehydrogenase (LDH) release and expression of matrix metalloproteinase-2 (MMP-2) and matrix metalloproteinase-9 (MMP-9). Furthermore, Opicapone alleviated homocysteine-induced decrease in claudin-2 level in bEnd.3 cells. In summary, our results show that Opicapone protects against HHcy-induced BBB permeability by reducing the expression and gelatinase activity of MMPs, and increasing the expression of claudin-2.
Collapse
Affiliation(s)
- Jian Wang
- Department of Neurology, Ya'an Peoples Hospital, Ya'an, 625000, Sichuan, China
| | - Bo Zheng
- Department of Neurology, Ya'an Peoples Hospital, Ya'an, 625000, Sichuan, China
| | - Shu Yang
- Department of Neurology, The Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, 610000, Sichuan, China
| | - Hui Zheng
- Department of Neurology, Chengdu First People's Hospital, Chengdu, 610000, Sichuan, China.
| | - Jianhong Wang
- Department of Neurology, The Affiliated Hospital of University of Electronic Science and Technology, Sichuan Provincial People's Hospital, Chengdu, 610000, Sichuan, China.
| |
Collapse
|
25
|
Cancer-associated fibroblasts as cellular vehicles in endometrial cancer cell migration. Oncol Lett 2021; 23:3. [PMID: 34820002 PMCID: PMC8607233 DOI: 10.3892/ol.2021.13121] [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] [Academic Contribution Register] [Received: 07/03/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022] Open
Abstract
Cell motility is a critical step in the metastasis cascade. However, the role of cancer-associated fibroblasts (CAFs) in facilitating endometrial cancer (EC) cell motility remains unclear. The present study aimed to investigate the role of CAFs in EC motility in a 3D environment. A co-culture model was established using an EC cell line (ECC-1) and CAFs on a Matrigel® matrix and compared to the respective individual monocultures. It was demonstrated that endometrial CAFs increased the motility of the EC cell line, compared with the monoculture. Using live cell imaging, CAFs were observed to form cell projections that served as contact guidance for ECC-1 cell locomotion in the spheroid formation process. These effects were specific to CAFs, as fibroblasts isolated from benign endometrial tissue samples did not form cell projections. Molecular analysis revealed that RhoA/Rho-associated, coiled-coil containing protein kinase 1 (ROCK1) signaling activation partly contributed to CAF-mediated ECC-1 cell migration. The presence of Matrigel® increased the mRNA expression of RhoA, and the mRNA and protein expression levels of its downstream effectors, ROCK1 and p-MLC, respectively, in the ECC-1 and CAF co-culture, as well as the ECC-1 and CAF monocultures. Interestingly, high phosphorylation levels of myosin light chain mediated the activation of RhoA/ROCK1 signaling in the ECC-1 and CAF co-culture. The ROCK1 inhibitor Y-27632 attenuated the motility of tumor cells in ECC-1 and CAF co-cultures. However, similar treatment led to a significant inhibition in the motility of the CAF monoculture, but not the ECC-1 monoculture. Moreover, tumor spheroid formation was inhibited due to a reduction in stress fiber formation in ECC-1 and CAF co-cultures. Altogether, these findings suggest that the regulation of the RhoA/ROCK1 signaling pathway is required for CAFs to serve as cellular vehicles in order for EC cells to migrate and form spheroids in a 3D environment.
Collapse
|
26
|
Li X, Tong J, Liu J, Wang Y. Down-regulation of ROCK2 alleviates ethanol-induced cerebral nerve injury partly by the suppression of the NF-κB signaling pathway. Bioengineered 2021; 11:779-790. [PMID: 32684089 PMCID: PMC8291877 DOI: 10.1080/21655979.2020.1795404] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/20/2022] Open
Abstract
Chronic alcohol consumption leads to hippocampal neuronal impairment, which related to neuronal death, oxidative stress, and inflammatory response. Rho-associated protein kinase 2 (ROCK2) is a major regulator in the central nervous system injury. However, the effects of ROCK2 in ethanol-induced brain injury have not been explored. In this work, we investigated the neuroprotective effects and the mechanism of ROCK2 inhibition in vivo. Wistar rats were exposed to 37% ethanol for 8 weeks to establish brain injury models. Morris water maze test was performed to evaluate cognitive function, and we found that the down-regulation of ROCK2 reduced the escape latency and increased the passing times and percentage of time spent in the target quadrant of rats. The results of H&E staining and Nissl staining showed that ROCK2 inhibition alleviated the pathological injury induced by ethanol. PI staining and Western blot confirmed that inhibiting ROCK2 attenuated the neuronal death and apoptosis as reflected by the reduced PI-positive neurons and the decreased expression of cleaved-caspase-3 and cleaved-caspase-9. Furthermore, the down-regulation of ROCK2 ameliorated the oxidative stress and inflammatory response induced by ethanol in rats as reflected by the up-regulation of IL-10, SOD, and GSH and reduction of TNF-α, IL-6, and MDA respectively. Additionally, Western blot and EMSA analysis revealed that the down-regulation of ROCK2 suppressed the nuclear transfer of NF-κB p65. In conclusion, our data suggested that ROCK2 inhibition ameliorated ethanol-mediated hippocampal neuronal impairment by anti-apoptotic, anti-inflammatory, anti-oxidative effects at least partially through the suppression of the NF-κB pathway.
Collapse
Affiliation(s)
- Xinguo Li
- Department of Neurosurgery, The First Hospital of China Medical University , Shenyang, People's Republic of China
| | - Jing Tong
- Department of Gastroenterology, The First Hospital of China Medical University , Shenyang, People's Republic of China
| | - Jihui Liu
- Department of Neurosurgery, The First Hospital of China Medical University , Shenyang, People's Republic of China
| | - Yibao Wang
- Department of Neurosurgery, The First Hospital of China Medical University , Shenyang, People's Republic of China
| |
Collapse
|
27
|
Stepankova K, Jendelova P, Machova Urdzikova L. Planet of the AAVs: The Spinal Cord Injury Episode. Biomedicines 2021; 9:613. [PMID: 34071245 PMCID: PMC8228984 DOI: 10.3390/biomedicines9060613] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/07/2021] [Revised: 05/22/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022] Open
Abstract
The spinal cord injury (SCI) is a medical and life-disrupting condition with devastating consequences for the physical, social, and professional welfare of patients, and there is no adequate treatment for it. At the same time, gene therapy has been studied as a promising approach for the treatment of neurological and neurodegenerative disorders by delivering remedial genes to the central nervous system (CNS), of which the spinal cord is a part. For gene therapy, multiple vectors have been introduced, including integrating lentiviral vectors and non-integrating adeno-associated virus (AAV) vectors. AAV vectors are a promising system for transgene delivery into the CNS due to their safety profile as well as long-term gene expression. Gene therapy mediated by AAV vectors shows potential for treating SCI by delivering certain genetic information to specific cell types. This review has focused on a potential treatment of SCI by gene therapy using AAV vectors.
Collapse
Affiliation(s)
- Katerina Stepankova
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská 1083, 14200 Prague, Czech Republic;
- Department of Neuroscience, Second Faculty of Medicine, Charles University, 15006 Prague, Czech Republic
| | - Pavla Jendelova
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská 1083, 14200 Prague, Czech Republic;
- Department of Neuroscience, Second Faculty of Medicine, Charles University, 15006 Prague, Czech Republic
| | - Lucia Machova Urdzikova
- Institute of Experimental Medicine, Czech Academy of Sciences, Vídeňská 1083, 14200 Prague, Czech Republic;
- Department of Neuroscience, Second Faculty of Medicine, Charles University, 15006 Prague, Czech Republic
| |
Collapse
|
28
|
Guiler W, Koehler A, Boykin C, Lu Q. Pharmacological Modulators of Small GTPases of Rho Family in Neurodegenerative Diseases. Front Cell Neurosci 2021; 15:661612. [PMID: 34054432 PMCID: PMC8149604 DOI: 10.3389/fncel.2021.661612] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/31/2021] [Accepted: 04/08/2021] [Indexed: 12/22/2022] Open
Abstract
Classical Rho GTPases, including RhoA, Rac1, and Cdc42, are members of the Ras small GTPase superfamily and play essential roles in a variety of cellular functions. Rho GTPase signaling can be turned on and off by specific GEFs and GAPs, respectively. These features empower Rho GTPases and their upstream and downstream modulators as targets for scientific research and therapeutic intervention. Specifically, significant therapeutic potential exists for targeting Rho GTPases in neurodegenerative diseases due to their widespread cellular activity and alterations in neural tissues. This study will explore the roles of Rho GTPases in neurodegenerative diseases with focus on the applications of pharmacological modulators in recent discoveries. There have been exciting developments of small molecules, nonsteroidal anti-inflammatory drugs (NSAIDs), and natural products and toxins for each classical Rho GTPase category. A brief overview of each category followed by examples in their applications will be provided. The literature on their roles in various diseases [e.g., Alzheimer's disease (AD), Parkinson's disease (PD), Amyotrophic lateral sclerosis (ALS), Frontotemporal dementia (FTD), and Multiple sclerosis (MS)] highlights the unique and broad implications targeting Rho GTPases for potential therapeutic intervention. Clearly, there is increasing knowledge of therapeutic promise from the discovery of pharmacological modulators of Rho GTPases for managing and treating these conditions. The progress is also accompanied by the recognition of complex Rho GTPase modulation where targeting its signaling can improve some aspects of pathogenesis while exacerbating others in the same disease model. Future directions should emphasize the importance of elucidating how different Rho GTPases work in concert and how they produce such widespread yet different cellular responses during neurodegenerative disease progression.
Collapse
Affiliation(s)
| | | | | | - Qun Lu
- Department of Anatomy and Cell Biology, The Harriet and John Wooten Laboratory for Alzheimer’s and Neurogenerative Diseases Research, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| |
Collapse
|
29
|
Batra M, Gupta S, Nair AB, Dhanawat M, Sandal S, Morsy MA. Netarsudil: A new ophthalmic drug in the treatment of chronic primary open angle glaucoma and ocular hypertension. Eur J Ophthalmol 2021; 31:2237-2244. [PMID: 33843288 DOI: 10.1177/11206721211008783] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Vision impairment remains a major health problem worldwide. Elevated intraocular pressure is a prime risk factor for blindness in the elderly. Netarsudil is a Rho-associated protein kinase (ROCK) inhibitor, which also inhibits norepinephrine transport. This narrative review summarizes the properties and clinical significance of netarsudil, a promising drug in topical glaucoma therapy. METHODS We searched PubMed, Medline and Scopus databases using relevant keywords to retrieve information on the physicochemical properties, formulation, mechanism of action, clinical pharmacokinetics, dose and toxicity of netarsudil. RESULTS Netarsudil showed promising effects in lowering the elevated intraocular pressure by two mechanisms. The US FDA approved netarsudil for clinical use in 2017 under the trademark of Rhopressa® while European Medicines Agency approved Rhokiinsa® in 2019. This drug is available as a 0.02% ophthalmic solution for once-daily topical application. CONCLUSION The discovery of netarsudil is a breakthrough in the therapy of glaucoma with proven efficacy in a wide range of eye pressures and is well tolerated in cases with ocular hypertension and chronic glaucoma.
Collapse
Affiliation(s)
- Mansi Batra
- Department of Clinical Practice, M. M. College of Pharmacy, M. M. (Deemed to be University), Mullana (Ambala), Haryana, India
| | - Sumeet Gupta
- Department of Pharmacology, M. M. College of Pharmacy, M. M. (Deemed to be University), Mullana (Ambala), Haryana, India
| | - Anroop B Nair
- College of Clinical Pharmacy, Department of Pharmaceutical Sciences, King Faisal University, Al-Ahsa, Saudi Arabia
| | - Meenakshi Dhanawat
- Department of Pharmaceutical Chemistry, M. M. College of Pharmacy, M. M. (Deemed to be University), Mullana (Ambala), Haryana, India
| | - Suraj Sandal
- Department of Pharmacology, M. M. College of Pharmacy, M. M. (Deemed to be University), Mullana (Ambala), Haryana, India
| | - Mohamed Aly Morsy
- College of Clinical Pharmacy, Department of Pharmaceutical Sciences, King Faisal University, Al-Ahsa, Saudi Arabia.,Department of Pharmacology, Faculty of Medicine, Minia University, El-Minia, Egypt
| |
Collapse
|
30
|
Ruan Z, Li Y, He R, Li X. Inhibition of microRNA-10b-5p up-regulates HOXD10 to attenuate Alzheimer's disease in rats via the Rho/ROCK signalling pathway. J Drug Target 2021; 29:531-540. [PMID: 33307856 DOI: 10.1080/1061186x.2020.1864739] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVE It is believed that microRNAs (miRNAs) participate in the pathogenesis of Alzheimer's disease (AD), but the specified function of miR-10b-5p in the disease has not been thoroughly understood. Thereafter, this research aimed to assess the function of miR-10b-5p in AD. METHODS Rat AD models were established by injected with amyloid-β1-42 (Aβ1-42), which were mainly treated with lentivirus-miR-10b-5p inhibitor, or lentivirus-overexpressed homeobox D10 (HOXD10). MiR-10b-5p, HOXD10, RhoA, ROCK1 and ROCK2 expression in rat hippocampal tissues were determined. Afterwards, the behaviour of rats was tested, and neuronal apoptosis, pathological injury, and inflammatory factors and oxidative stress-related factors were all assessed. Finally, the target relation between miR-10b-5p and HOXD10 was detected. RESULTS MiR-10b-5p was upregulated while HOXD10 was downregulated, and the Rho/ROCK signalling pathway was activated in hippocampal tissues of rats with AD. Inhibition of miR-10b-5p could attenuate the neuronal apoptosis, pathological injury, inflammation reaction, and oxidative stress by elevating HOXD10 and inhibiting the Rho/ROCK signalling pathway in AD rats. Moreover, HOXD10 was targeted by miR-10b-5p. CONCLUSION Inhibited miR-10b-5p decelerated the development of AD by promoting HOXD10 and inactivating the Rho/ROCK signalling pathway, and our findings may contribute to the exploration of AD treatment.
Collapse
Affiliation(s)
- Zhongfan Ruan
- Department of Neurology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| | - Yan Li
- Department of Anesthesiology, Affiliated Nanhua Hospital, University of South China, Hengyang, Hunan, China
| | - Rongzhang He
- Translational Medicine Institute, National & Local Joint Engineering Laboratory for High-through Molecular Diagnosis Technology, Collaborative Research Center for Post-doctoral Mobile Stations of Central South University, Affiliated the First People's Hospital of Chenzhou of University of South China, Chenzhou, Hunan, China
| | - Xuewei Li
- Department of Neurology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, China
| |
Collapse
|
31
|
Mukherjee N, Adak A, Ghosh S. Recent trends in the development of peptide and protein-based hydrogel therapeutics for the healing of CNS injury. SOFT MATTER 2020; 16:10046-10064. [PMID: 32724981 DOI: 10.1039/d0sm00885k] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 06/11/2023]
Abstract
Traumatic brain injury (TBI) and spinal cord injury (SCI) cause millions of deaths and permanent or prolonged physical disabilities around the globe every year. It generally happens due to various incidents, such as accidents during sports, war, physical assault, and strokes which result in severe damage to brain and spinal cord. If this remains untreated, traumatic CNS injuries may lead to early development of several neurodegenerative diseases like Alzheimer's, Parkinson, multiple sclerosis, and other mental illnesses. The initial physical reaction, which is also termed as the primary phase, includes swelling, followed by inflammation as a result of internal haemorrhage causing damage to indigenous tissue, i.e., axonal shear injury, rupture of blood vessels, and partial impaired supply of oxygen and essential nutrients in the neurons, thereby initiating a cascade of events causing secondary injuries such as hypoxia, hypotension, cognitive impairment, seizures, imbalanced calcium homeostasis and glutamate-induced excitotoxicity resulting in concomitant neuronal cell death and cumulative permanent tissue damage. In the modern era of advanced biomedical technology, we are still living with scarcity of the clinically applicable comparative non-invasive therapeutic strategies for regeneration or functional recovery of neurons or neural networks after a massive CNS injury. One of the key reasons for this scarcity is the limited regenerative ability of neurons in CNS. Growth-impermissive glial scar and the lack of a synthetic biocompatible platform for proper neural tissue engineering and controlled supply of drugs further retard the healing process. Injectable or implantable hydrogel materials, consisting majorly of water in its porous three-dimensional (3D) structure, can serve as an excellent drug delivery platform as well as a transplanted cell-supporting scaffold medium. Among the various neuro-compatible bioinspired materials, we are limiting our discussion to the recent advancement of engineered biomaterials comprising mainly of peptides and proteins due to their growing demand, low immunogenicity and versatility in the fabrication of neuro regenerative medicine. In this article, we try to explore all the recent scientific avenues that are developing gradually to make peptide and peptide-conjugated biomaterial hydrogels as a therapeutic and supporting scaffold for treating CNS injuries.
Collapse
Affiliation(s)
- Nabanita Mukherjee
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur, NH 65, Surpura Bypass Road, Karwar, Rajasthan 342037, India.
| | | | | |
Collapse
|
32
|
Extracellular Matrix in Neural Plasticity and Regeneration. Cell Mol Neurobiol 2020; 42:647-664. [PMID: 33128689 DOI: 10.1007/s10571-020-00986-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/30/2020] [Accepted: 10/22/2020] [Indexed: 12/19/2022]
Abstract
The extracellular matrix (ECM) is a fundamental component of biological tissues. The ECM in the central nervous system (CNS) is unique in both composition and function. Functions such as learning, memory, synaptogenesis, and plasticity are regulated by numerous ECM molecules. The neural ECM acts as a non-specific physical barrier that modulates neuronal plasticity and axon regeneration. There are two specialized types of ECM in the CNS, diffuse perisynaptic ECM and condensed ECM, which selectively surround the perikaryon and initial part of dendritic trees in subtypes of neurons, forming perineuronal nets. This review presents the current knowledge about the role of important neuronal ECM molecules in maintaining the basic functions of a neuron, including electrogenesis and the ability to form neural circuits. The review mainly focuses on the role of ECM components that participate in the control of key events such as cell survival, axonal growth, and synaptic remodeling. Particular attention is drawn to the numerous molecular partners of the main ECM components. These regulatory molecules are integrated into the cell membrane or disposed into the matrix itself in solid or soluble form. The interaction of the main matrix components with molecular partners seems essential in molecular mechanisms controlling neuronal functions. Special attention is paid to the chondroitin sulfate proteoglycan 4, type 1 transmembrane protein, neural-glial antigen 2 (NG2/CSPG4), whose cleaved extracellular domain is such a molecular partner that it not only acts directly on neural and vascular cells, but also exerts its influence indirectly by binding to resident ECM molecules.
Collapse
|
33
|
Context-Dependent Role of miR-124 in Retinoic Acid-Induced Growth Cone Attraction of Regenerating Motorneurons. Cell Mol Neurobiol 2020; 42:847-869. [PMID: 33094464 DOI: 10.1007/s10571-020-00982-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/25/2020] [Accepted: 10/14/2020] [Indexed: 10/23/2022]
Abstract
During development and regeneration, growth cones at the tips of extending axons navigate through a complex environment to establish accurate connections with appropriate targets. Growth cones can respond rapidly to classical and non-classical guidance cues in their environment, often requiring local protein synthesis. In vertebrate growth cones, local protein synthesis in response to classical cues can require regulation by microRNAs (miRNAs), a class of small, conserved, non-coding RNAs that post-transcriptionally regulate gene expression. However, less is known of how miRNAs mediate growth cone responses to non-classical cues (such as retinoic acid (RA)), specifically in invertebrates. Here, we utilized adult regenerating invertebrate motorneurons to study miRNA regulation of growth cone attraction to RA, shown to require local protein synthesis. In situ hybridization revealed the presence of miR-124 in growth cones of regenerating ciliary motorneurons of the mollusc Lymnaea stagnalis. Changes in the spatiotemporal distribution of miR-124 occurred following application of RA, and dysregulation of miR-124 (with mimic injection), disrupted RA-induced growth cone turning in a time-dependent manner. This behavioural regulation by miR-124 was altered when the neurite was transected, and the growth cone completely separated from the soma. miR-124 did not, however, appear to be involved in growth cone attraction to serotonin, a response independent of local protein synthesis. Finally, we provide evidence that a downstream effector of RhoGTPases, ROCK, is a potential target of miR-124 during RA-induced growth cone responses. These data advance our current understanding of how microRNAs might mediate cue- and context-dependent behaviours during axon guidance.
Collapse
|
34
|
Zhao Y, Ge X, Yu H, Kuil LE, Alves MM, Tian D, Huang Q, Chen X, Hofstra RMW, Gao Y. Inhibition of ROCK signaling pathway accelerates enteric neural crest cell-based therapy after transplantation in a rat hypoganglionic model. Neurogastroenterol Motil 2020; 32:e13895. [PMID: 32515097 DOI: 10.1111/nmo.13895] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 11/25/2019] [Revised: 04/23/2020] [Accepted: 05/05/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Hirschsprung's disease (HSCR) is a congenital gastrointestinal disorder, characterized by enteric ganglia absence in part or entire of the colon, due to abnormal colonization and migration of enteric neural crest cells (ENCCs) during development. Currently, besides surgery which is the main therapy for HSCR, the potential of stem cell-based transplantation was investigated as an alternative option. Although promising, it has limitations, including poor survival, differentiation, and migration of the grafted cells. We hypothesized that modulation of extracellular factors during transplantation could promote ENCCs proliferation and migration, leading to increased transplantation efficiency. Considering that the RhoA/ROCK pathway is highly involved in cytoskeletal dynamics and neurite growth, our study explored the effect of inhibition of this pathway to improve the success of ENCCs transplantation. METHODS Enteric neural crest cells were isolated from rat embryos and labeled with a GFP-tag. Cell viability, apoptosis, differentiation, and migration assays were performed with and without RhoA/ROCK inhibition. Labeled ENCCs were transplanted into the muscle layer of an induced hypoganglionic rat model followed by intraperitoneal injections of ROCK inhibitor. The transplanted segments were collected 3 weeks after for histological analysis. KEY RESULTS Our results showed that inhibition of ROCK increased viable cell number, differentiation, and migration of ENCCs in vitro. Moreover, transplantation of labeled ENCCs into the hypoganglionic model showed enhanced distribution of grafted ENCCs, upon treatment with ROCK inhibitor. CONCLUSIONS AND INFERENCES ROCK inhibitors influence ENCCs growth and migration in vitro and in vivo, and should be considered to improve the efficiency of ENCCs transplantation.
Collapse
Affiliation(s)
- Yuying Zhao
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.,Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Xin Ge
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hui Yu
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Laura E Kuil
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Maria M Alves
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Donghao Tian
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qiang Huang
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xinlin Chen
- Institute of Neurobiology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China
| | - Robert M W Hofstra
- Department of Clinical Genetics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Ya Gao
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
35
|
Matoba K, Takeda Y, Nagai Y, Kanazawa Y, Kawanami D, Yokota T, Utsunomiya K, Nishimura R. ROCK Inhibition May Stop Diabetic Kidney Disease. JMA J 2020; 3:154-163. [PMID: 33150249 PMCID: PMC7590381 DOI: 10.31662/jmaj.2020-0014] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/05/2020] [Accepted: 03/31/2020] [Indexed: 12/20/2022] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of end-stage renal disease and is strongly associated with cardiovascular mortality. Given the pandemic of obesity and diabetes, the elucidation of the molecular underpinnings of DKD and establishment of effective therapy are urgently required. Studies over the past decade have identified the activated renin-angiotensin system (RAS) and hemodynamic changes as important therapeutic targets. However, given the residual risk observed in patients treated with RAS inhibitors and/or sodium glucose co-transporter 2 inhibitors, the involvement of other molecular machinery is likely, and the elucidation of such pathways represents fertile ground for the development of novel strategies. Rho-kinase (ROCK) is a serine/threonine kinase that is under the control of small GTPase protein Rho. Many fundamental cellular processes, including migration, proliferation, and survival are orchestrated by ROCK through a mechanism involving cytoskeletal reorganization. From a pathological standpoint, several analyses provide compelling evidence supporting the hypothesis that ROCK is an important regulator of DKD that is highly pertinent to cardiovascular disease. In cell-based studies, ROCK is activated in response to a diverse array of external stimuli associated with diabetes, and renal ROCK activity is elevated in the context of type 1 and 2 diabetes. Experimental studies have demonstrated the efficacy of pharmacological or genetic inhibition of ROCK in the prevention of diabetes-related histological and functional abnormalities in the kidney. Through a bird’s eye view of ROCK in renal biology, the present review provides a conceptual framework that may be widely applicable to the pathological processes of multiple organs and illustrate novel therapeutic promise in diabetology.
Collapse
Affiliation(s)
- Keiichiro Matoba
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yusuke Takeda
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yosuke Nagai
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yasushi Kanazawa
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Daiji Kawanami
- Department of Endocrinology and Diabetes Mellitus, Fukuoka University School of Medicine, Fukuoka, Japan
| | - Tamotsu Yokota
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Kazunori Utsunomiya
- Center for Preventive Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Rimei Nishimura
- Division of Diabetes, Metabolism, and Endocrinology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
36
|
Stem Cell Therapy for Neurogenic Bladder After Spinal Cord Injury: Clinically Possible? Int Neurourol J 2020; 24:S3-10. [PMID: 32482052 PMCID: PMC7285699 DOI: 10.5213/inj.2040150.075] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/01/2020] [Accepted: 05/09/2020] [Indexed: 12/29/2022] Open
Abstract
Neurogenic bladder (NB) after spinal cord injury (SCI) is a common complication that inhibits normal daily activities and reduces the quality of life. Regrettably, the current therapeutic methods for NB are inadequate. Therefore, numerous studies have been conducted to develop new treatments for NB associated with SCI. Moreover, a myriad of preclinical and clinical trials on the effects and safety of stem cell therapy in patients with SCI have been performed, and several studies have demonstrated improvements in urodynamic parameters, as well as in sensory and motor function, after stem cell therapy. These results are promising; however, further high-quality clinical studies are necessary to compensate for a lack of randomized trials, the modest number of participants, variation in the types of stem cells used, and inconsistency in routes of administration.
Collapse
|
37
|
Rigby MJ, Gomez TM, Puglielli L. Glial Cell-Axonal Growth Cone Interactions in Neurodevelopment and Regeneration. Front Neurosci 2020; 14:203. [PMID: 32210757 PMCID: PMC7076157 DOI: 10.3389/fnins.2020.00203] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/20/2020] [Accepted: 02/24/2020] [Indexed: 12/19/2022] Open
Abstract
The developing nervous system is a complex yet organized system of neurons, glial support cells, and extracellular matrix that arranges into an elegant, highly structured network. The extracellular and intracellular events that guide axons to their target locations have been well characterized in many regions of the developing nervous system. However, despite extensive work, we have a poor understanding of how axonal growth cones interact with surrounding glial cells to regulate network assembly. Glia-to-growth cone communication is either direct through cellular contacts or indirect through modulation of the local microenvironment via the secretion of factors or signaling molecules. Microglia, oligodendrocytes, astrocytes, Schwann cells, neural progenitor cells, and olfactory ensheathing cells have all been demonstrated to directly impact axon growth and guidance. Expanding our understanding of how different glial cell types directly interact with growing axons throughout neurodevelopment will inform basic and clinical neuroscientists. For example, identifying the key cellular players beyond the axonal growth cone itself may provide translational clues to develop therapeutic interventions to modulate neuron growth during development or regeneration following injury. This review will provide an overview of the current knowledge about glial involvement in development of the nervous system, specifically focusing on how glia directly interact with growing and maturing axons to influence neuronal connectivity. This focus will be applied to the clinically-relevant field of regeneration following spinal cord injury, highlighting how a better understanding of the roles of glia in neurodevelopment can inform strategies to improve axon regeneration after injury.
Collapse
Affiliation(s)
- Michael J Rigby
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, United States.,Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, United States.,Waisman Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Timothy M Gomez
- Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, United States.,Department of Neuroscience, University of Wisconsin-Madison, Madison, WI, United States
| | - Luigi Puglielli
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, United States.,Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, United States.,Waisman Center, University of Wisconsin-Madison, Madison, WI, United States.,Geriatric Research Education Clinical Center, Veterans Affairs Medical Center, Madison, WI, United States
| |
Collapse
|
38
|
Tang Y, Han L, Bai X, Liang X, Zhao J, Huang F, Wang J. Intranasal Delivery of Bone Marrow Stromal Cells Preconditioned with Fasudil to Treat a Mouse Model of Parkinson's Disease. Neuropsychiatr Dis Treat 2020; 16:249-262. [PMID: 32158210 PMCID: PMC6986408 DOI: 10.2147/ndt.s238646] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 11/14/2019] [Accepted: 01/13/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Stem cell transplantation is a promising strategy with great potential to treat Parkinson's disease (PD). Nevertheless, improving the cell delivery route and optimising implanted cells are necessary to increase the therapeutic effect. Herein, we investigated whether intranasal delivery of bone marrow stromal cells (BMSCs) has beneficial effects in a PD mouse model and whether the therapeutic potential of BMSCs could be enhanced by preconditioning with fasudil. METHODS A PD mouse model was developed by intraperitoneally administering 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Mice were treated intranasally with phosphate buffered saline (PBS), BMSCs, or BMSCs preconditioned with fasudil. One month later, the effects of BMSC treatment were analysed. RESULTS Our study showed that fasudil could accelerate the proliferation of BMSCs and promote brain-derived neurotrophic factor (BDNF) secretion in vitro. Intranasally administered BMSCs were capable of surviving and migrating in the brain. Intranasal delivery of BMSCs preconditioned with fasudil significantly improved motor function and reduced dopaminergic neuron loss in substantia nigra; treatment with BMSCs and PBS resulted in similar outcomes. Preconditioning with fasudil inhibited the activation and aggregation of microglia, suppressed immune response, and reinforced BDNF secretion in MPTP-PD mice significantly more than treatment with BMSCs alone. CONCLUSION The present study demonstrates that intranasally administering BMSCs preconditioned with fasudil is a promising cell-based therapy for PD.
Collapse
Affiliation(s)
- Yilin Tang
- Department of Neurology and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China
| | - Linlin Han
- Department of Neurology and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China
| | - Xiaochen Bai
- Department of Neurology and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China.,The State Key Laboratory of Medical Neurobiology, The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Xiaoniu Liang
- Department of Neurology and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China
| | - Jue Zhao
- Department of Neurology and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China
| | - Fang Huang
- The State Key Laboratory of Medical Neurobiology, The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, People's Republic of China
| | - Jian Wang
- Department of Neurology and National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, People's Republic of China
| |
Collapse
|
39
|
Nimgampalle M, Banavath HN, Chakravarthy H, Saxena A, Devanathan V. Novel inhibitors of Rho-kinase mediated neuroinflammatory pathways and their potential application in recovery of injured spinal cord. J Biomol Struct Dyn 2019; 38:4669-4686. [DOI: 10.1080/07391102.2019.1686066] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/27/2022]
Affiliation(s)
- Mallikarjuna Nimgampalle
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, Andhra Pradesh, India
| | - Hemanth Naick Banavath
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, Andhra Pradesh, India
| | - Harshini Chakravarthy
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, Andhra Pradesh, India
| | | | - Vasudharani Devanathan
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati, Andhra Pradesh, India
| |
Collapse
|
40
|
Wang L, Xu J, Guo D, Zhou X, Jiang W, Wang J, Tang J, Zou Y, Bi M, Li Q. Fasudil alleviates brain damage in rats after carbon monoxide poisoning through regulating neurite outgrowth inhibitor/oligodendrocytemyelin glycoprotein signalling pathway. Basic Clin Pharmacol Toxicol 2019; 125:152-165. [PMID: 30916885 DOI: 10.1111/bcpt.13233] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/29/2018] [Accepted: 02/07/2019] [Indexed: 12/21/2022]
Abstract
Carbon monoxide (CO) poisoning can lead to many serious neurological symptoms. Currently, there are no effective therapies for CO poisoning. In this study, rats exposed to CO received hyperbaric oxygen therapy, and those in the Fasudil group were given additional Fasudil injection once daily. We found that the escape latency in CO poisoning group (CO group) was significantly prolonged, the T1 /Ttotal was obviously decreased, and the mean escape time and the active escape latency were notably extended compared with those in normal control group (NC group, P < 0.05). After administration of Fasudil, the escape latency was significantly shortened, T1 /Ttotal was gradually increased as compared with CO group (>1 week, P < 0.05). Ultrastructural damage of neurons and blood-brain barrier of rats was serious in CO group, while the structural and functional integrity of neuron and mitochondria maintained relatively well in Fasudil group. Moreover, we also noted that the expressions of neurite outgrowth inhibitor (Nogo), oligodendrocyte-myelin glycoprotein (OMgp) and Rock in brain tissue were significantly increased in CO group, and the elevated levels of the three proteins were still observed at 2 months after CO poisoning. Fasudil markedly reduced their expressions compared with those of CO group (P < 0.05). In summary, the activation of Nogo-OMgp/Rho signalling pathway is associated with brain injury in rats with CO poisoning. Fasudil can efficiently down-regulate the expressions of Nogo, OMgp and Rock proteins, paving a way for the treatment of acute brain damage after CO poisoning.
Collapse
Affiliation(s)
- Li Wang
- Department of Neurology, Qianfoshan Hospital Affiliated to Shandong University, Jinan Shandong, China.,Department of Integration of Chinese and Western Medicine, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai Shandong, China
| | - Jianghua Xu
- Department of neurology, Yantai YEDA Hospital, Yantai Shandong, China
| | - Dadong Guo
- Eye Institute of Shandong University of Traditional Chinese Medicine, Jinan Shandong, China
| | - Xudong Zhou
- The First Affiliated Hospital of Shandong, University of Traditional Chinese Medicine, Jinan Shandong, China
| | - Wenwen Jiang
- Department of Integration of Chinese and Western Medicine, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai Shandong, China
| | - Jinglin Wang
- Emergency Centre, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai Shandong, China
| | - Jiyou Tang
- Department of Neurology, Qianfoshan Hospital Affiliated to Shandong University, Jinan Shandong, China
| | - Yong Zou
- Department of Integration of Chinese and Western Medicine, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai Shandong, China
| | - Mingjun Bi
- Emergency Centre, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai Shandong, China
| | - Qin Li
- Department of Integration of Chinese and Western Medicine, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai Shandong, China
| |
Collapse
|
41
|
Sun XZ, Li SY, Tian XY, Hong Z, Li JX. Effect of Rho kinase inhibitor fasudil on the expression ET-1 and NO in rats with hypoxic pulmonary hypertension. Clin Hemorheol Microcirc 2019; 71:3-8. [PMID: 29660902 DOI: 10.3233/ch-160232] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/19/2022]
Affiliation(s)
- Xing-Zhen Sun
- Department of Pediatrics, Huai’an First People’s Hospital, Nanjing Medical University, Huai’an, China
| | - Shu-Yan Li
- Department of Ophthalmology, Huai’an First People’s Hospital, Nanjing Medical University, Huai’an, China
| | - Xiang-Yang Tian
- Department of Neurology, Huai’an First People’s Hospital, Nanjing Medical University, Huai’an, China
| | - Ze Hong
- Department of Pediatrics, Huai’an First People’s Hospital, Nanjing Medical University, Huai’an, China
| | - Jia-Xin Li
- Department of Pediatrics, Huai’an First People’s Hospital, Nanjing Medical University, Huai’an, China
| |
Collapse
|
42
|
Tanna AP, Johnson M. Rho Kinase Inhibitors as a Novel Treatment for Glaucoma and Ocular Hypertension. Ophthalmology 2018; 125:1741-1756. [PMID: 30007591 PMCID: PMC6188806 DOI: 10.1016/j.ophtha.2018.04.040] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/13/2018] [Revised: 04/24/2018] [Accepted: 04/30/2018] [Indexed: 01/11/2023] Open
Abstract
In an elegant example of bench-to-bedside research, a hypothesis that cells in the outflow pathway actively regulate conventional outflow resistance was proposed in the 1990s and systematically pursued, exposing novel cellular and molecular mechanisms of intraocular pressure (IOP) regulation. The critical discovery that pharmacologic manipulation of the cytoskeleton of outflow pathway cells decreased outflow resistance placed a spotlight on the Rho kinase pathway that was known to regulate the cytoskeleton. Ultimately, a search for Rho kinase inhibitors led to the discovery of several molecules of therapeutic interest, leaving us today with 2 new ocular hypotensive agents approved for clinical use: ripasudil in Japan and netarsudil in the United States. These represent members of the first new class of clinically useful ocular hypotensive agents since the US Food and Drug Administration approval of latanoprost in 1996. The development of Rho kinase inhibitors as a class of medications to lower IOP in patients with glaucoma and ocular hypertension represents a triumph in translational research. Rho kinase inhibitors are effective alone or when combined with other known ocular hypotensive medications. They also offer the possibility of neuroprotective activity, a favorable impact on ocular blood flow, and even an antifibrotic effect that may prove useful in conventional glaucoma surgery. Local adverse effects, however, including conjunctival hyperemia, subconjunctival hemorrhages, and cornea verticillata, are common. Development of Rho kinase inhibitors targeted to the cells of the outflow pathway and the retina may allow these agents to have even greater clinical impact. The objectives of this review are to describe the basic science underlying the development of Rho kinase inhibitors as a therapy to lower IOP and to summarize the results of the clinical studies reported to date. The neuroprotective and vasoactive properties of Rho kinase inhibitors, as well as the antifibrotic properties, of these agents are reviewed in the context of their possible role in the medical and surgical treatment of glaucoma.
Collapse
Affiliation(s)
- Angelo P Tanna
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| | - Mark Johnson
- Department of Ophthalmology, Northwestern University Feinberg School of Medicine, Chicago, Illinois; Department of Biomedical Engineering, Northwestern University, Evanston, Illinois; Department of Mechanical Engineering, Northwestern University, Evanston, Illinois
| |
Collapse
|
43
|
Mertsch S, Schlicht K, Melkonyan H, Schlatt S, Thanos S. snRPN controls the ability of neurons to regenerate axons. Restor Neurol Neurosci 2018; 36:31-43. [PMID: 29439367 DOI: 10.3233/rnn-170780] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Retinal ganglion cells (RGCs) of mammals lose the ability to regenerate injured axons during postnatal maturation, but little is known about the underlying molecular mechanisms. OBJECTIVE It remains of particular importance to understand the mechanisms of axonal regeneration to develop new therapeutic approaches for nerve injuries. METHODS Retinas from newborn to adult monkeys (Callithrix jacchus)1 were obtained immediately after death and cultured in vitro. Growths of axons were monitored using microscopy and time-lapse video cinematography. Immunohistochemistry, Western blotting, qRT-PCR, and genomics were performed to characterize molecules associated with axonal regeneration and growth. A genomic screen was performed by using retinal explants versus native and non-regenerative explants obtained from eye cadavers on the day of birth, and hybridizing the mRNA with cross-reacting cDNA on conventional human microarrays. Followed the genomic screen, siRNA experiments were conducted to identify the functional involvement of identified candidates. RESULTS Neuron-specific human ribonucleoprotein N (snRPN) was found to be a potential regulator of impaired axonal regeneration during neuronal maturation in these animals. In particular, up-regulation of snRPN was observed during retinal maturation, coinciding with a decline in regenerative ability. Axon regeneration was reactivated in snRPN-knockout retinal ex vivo explants of adult monkey. CONCLUSION These results suggest that coordinated snRPN-driven activities within the neuron-specific ribonucleoprotein complex regulate the regenerative ability of RGCs in primates, thereby highlighting a potential new role for snRPN within neurons and the possibility of novel postinjury therapies.
Collapse
Affiliation(s)
- Sonja Mertsch
- Institute of Experimental Ophthalmology and DFG-Excellence Center, Cells in Motion (CiM, area C.4), School of Medicine, University of Münster, Münster, Germany.,Department of Ophthalmology, Laboratory of Experimental Ophthalmology, University Clinic Duesseldorf, Duesseldorf, Germany
| | - Katrin Schlicht
- Institute of Experimental Ophthalmology and DFG-Excellence Center, Cells in Motion (CiM, area C.4), School of Medicine, University of Münster, Münster, Germany
| | - Harutyun Melkonyan
- Institute of Experimental Ophthalmology and DFG-Excellence Center, Cells in Motion (CiM, area C.4), School of Medicine, University of Münster, Münster, Germany
| | - Stefan Schlatt
- Institute of Regenerative Medicine (CeRA) and DFG-Excellence Center, Cells in Motion (CiM, area A.2), School of Medicine, University of Münster, Münster, Germany
| | - Solon Thanos
- Institute of Experimental Ophthalmology and DFG-Excellence Center, Cells in Motion (CiM, area C.4), School of Medicine, University of Münster, Münster, Germany
| |
Collapse
|
44
|
Gunaydin C, Bilge SS. Effects of Nonsteroidal Anti-Inflammatory Drugs at the Molecular Level. Eurasian J Med 2018; 50:116-121. [PMID: 30002579 DOI: 10.5152/eurasianjmed.2018.0010] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/23/2017] [Accepted: 04/19/2018] [Indexed: 01/07/2023] Open
Abstract
Nonsteroidal anti-inflammatory drugs (NSAIDs) are commonly used for their anti-inflammatory, analgesic, and antipyretic effects. NSAIDs generally work by blocking the production of prostaglandins (PGs) through the inhibition of two cyclooxygenase enzymes. PGs are key factors in many cellular processes, such as gastrointestinal cytoprotection, hemostasis and thrombosis, inflammation, renal hemodynamics, turnover of cartilage, and angiogenesis. Interest has grown in the various effects of NSAIDs during the last decade. Epidemiological studies have revealed the reduced risk of several cancer types and neurodegenerative diseases by prolonged use of NSAIDs. Recent advances in the understanding of the cellular and molecular mechanisms of NSAIDs will accelerate the processes of discovery and clinical implementation. This review summarizes the molecular mechanisms of NSAIDs on the body systems.
Collapse
Affiliation(s)
- Caner Gunaydin
- Department of Pharmacology, Ondokuz Mayıs University School of Medicine, Samsun, Turkey
| | - S Sirri Bilge
- Department of Pharmacology, Ondokuz Mayıs University School of Medicine, Samsun, Turkey
| |
Collapse
|
45
|
Zhang G, Hu J, Rodemer W, Li S, Selzer ME. RhoA activation in axotomy-induced neuronal death. Exp Neurol 2018; 306:76-91. [PMID: 29715475 DOI: 10.1016/j.expneurol.2018.04.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/28/2017] [Revised: 04/18/2018] [Accepted: 04/27/2018] [Indexed: 01/11/2023]
Abstract
After spinal cord injury (SCI) in mammals, severed axons fail to regenerate, due to both extrinsic inhibitory factors, e.g., the chondroitin sulfate proteoglycans (CSPGs) and myelin-associated growth inhibitors (MAIs), and a developmental loss of intrinsic growth capacity. The latter is suggested by findings in lamprey that the 18 pairs of individually identified reticulospinal neurons vary greatly in their ability to regenerate their axons through the same spinal cord environment. Moreover, those neurons that are poor regenerators undergo very delayed apoptosis, and express common molecular markers after SCI. Thus the signaling pathways for retrograde cell death might converge with those inhibiting axon regeneration. Many extrinsic growth-inhibitory molecules activate RhoA, whereas inhibiting RhoA enhances axon growth. Whether RhoA also is involved in retrograde neuronal death after axotomy is less clear. Therefore, we cloned lamprey RhoA and correlated its mRNA expression and activation state with apoptosis signaling in identified reticulospinal neurons. RhoA mRNA was expressed widely in normal lamprey brain, and only slightly more in poorly-regenerating neurons than in good regenerators. However, within a day after spinal cord transection, RhoA mRNA was found in severed axon tips. Beginning at 5 days post-SCI RhoA mRNA was upregulated selectively in pre-apoptotic neuronal perikarya, as indicated by labelling with fluorescently labeled inhibitors of caspase activation (FLICA). After 2 weeks post-transection, RhoA expression decreased in the perikarya, and was translocated anterogradely into the axons. More striking than changes in RhoA mRNA levels, RhoA was continuously active selectively in FLICA-positive neurons through 9 weeks post-SCI. At that time, almost no neurons whose axons had regenerated were FLICA-positive. These findings are consistent with a role for RhoA activation in triggering retrograde neuronal death after SCI, and suggest that RhoA may be a point of convergence for inhibition of both axon regeneration and neuronal survival after axotomy.
Collapse
Affiliation(s)
- Guixin Zhang
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), USA
| | - Jianli Hu
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), USA
| | - William Rodemer
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), USA
| | - Shuxin Li
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), USA; Dept. Anatomy and Cell Biology, The Lewis Katz School of Medicine at Temple University, 3500 North Broad Street, Philadelphia, PA 19140, USA
| | - Michael E Selzer
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), USA; Dept. of Neurology, USA.
| |
Collapse
|
46
|
Nizamudeen ZA, Chakrabarti L, Sottile V. Exposure to the ROCK inhibitor fasudil promotes gliogenesis of neural stem cells in vitro. Stem Cell Res 2018; 28:75-86. [DOI: 10.1016/j.scr.2018.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 12/06/2017] [Revised: 01/30/2018] [Accepted: 02/02/2018] [Indexed: 12/27/2022] Open
|
47
|
Acute spinal cord injury: A review of pathophysiology and potential of non-steroidal anti-inflammatory drugs for pharmacological intervention. J Chem Neuroanat 2018; 87:25-31. [DOI: 10.1016/j.jchemneu.2017.08.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/06/2017] [Revised: 08/07/2017] [Accepted: 08/07/2017] [Indexed: 12/21/2022]
|
48
|
Fu PC, Tang RH, Yu ZY, Xie MJ, Wang W, Luo X. The Rho-associated kinase inhibitors Y27632 and fasudil promote microglial migration in the spinal cord via the ERK signaling pathway. Neural Regen Res 2018; 13:677-683. [PMID: 29722320 PMCID: PMC5950678 DOI: 10.4103/1673-5374.230294] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/30/2022] Open
Abstract
Rho-associated kinase (ROCK) is a key regulatory protein involved in inflammatory secretion in microglia in the central nervous system. Our previous studies showed that ROCK inhibition enhances phagocytic activity in microglia through the extracellular signal-regulated kinase (ERK) signaling pathway, but its effect on microglial migration was unknown. Therefore, in this study, we investigated the effects of the ROCK inhibitors Y27632 and fasudil on the migratory activity of primary cultured microglia isolated from the spinal cord, and we examined the underlying mechanisms. The microglia were treated with Y27632, fasudil and/or the ERK inhibitor U0126. Cellular morphology was observed by immunofluorescence. Transwell chambers were used to assess cell migration. ERK levels were measured by in-cell western blot assay. Y27632 and fasudil increased microglial migration, and the microglia were irregularly shaped and had many small processes. These inhibitors also upregulated the levels of phosphorylated ERK protein. The ERK inhibitor U0126 suppressed these effects of Y27632 and fasudil. These findings suggest that the ROCK inhibitors Y27632 and fasudil promote microglial migration in the spinal cord through the ERK signaling pathway.
Collapse
Affiliation(s)
- Pei-Cai Fu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Rong-Hua Tang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Zhi-Yuan Yu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Neurological Diseases (Huazhong University of Science and Technology), Ministry of Education of China, Wuhan, Hubei Province, China
| | - Min-Jie Xie
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Neurological Diseases (Huazhong University of Science and Technology), Ministry of Education of China, Wuhan, Hubei Province, China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Neurological Diseases (Huazhong University of Science and Technology), Ministry of Education of China, Wuhan, Hubei Province, China
| | - Xiang Luo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
49
|
A Ser75-to-Asp phospho-mimicking mutation in Src accelerates ageing-related loss of retinal ganglion cells in mice. Sci Rep 2017; 7:16779. [PMID: 29196663 PMCID: PMC5711949 DOI: 10.1038/s41598-017-16872-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/28/2017] [Accepted: 11/18/2017] [Indexed: 11/08/2022] Open
Abstract
Src knockout mice show no detectable abnormalities in central nervous system (CNS) post-mitotic neurons, likely reflecting functional compensation by other Src family kinases. Cdk1- or Cdk5-dependent Ser75 phosphorylation in the amino-terminal Unique domain of Src, which shares no homology with other Src family kinases, regulates the stability of active Src. To clarify the roles of Src Ser75 phosphorylation in CNS neurons, we established two types of mutant mice with mutations in Src: phospho-mimicking Ser75Asp (SD) and non-phosphorylatable Ser75Ala (SA). In ageing SD/SD mice, retinal ganglion cell (RGC) number in whole retinas was significantly lower than that in young SD/SD mice in the absence of inflammation and elevated intraocular pressure, resembling the pathogenesis of progressive optic neuropathy. By contrast, SA/SA mice and wild-type (WT) mice exhibited no age-related RGC loss. The age-related retinal RGC number reduction was greater in the peripheral rather than the mid-peripheral region of the retina in SD/SD mice. Furthermore, Rho-associated kinase activity in whole retinas of ageing SD/SD mice was significantly higher than that in young SD/SD mice. These results suggest that Src regulates RGC survival during ageing in a manner that depends on Ser75 phosphorylation.
Collapse
|
50
|
Mahmood J, Connors CQ, Alexander AA, Pavlovic R, Samanta S, Soman S, Matsui H, Sopko NA, Bivalacqua TJ, Weinreich D, Ho CY, Eley J, Sawant A, Jackson IL, Vujaskovic Z. Cavernous Nerve Injury by Radiation Therapy May Potentiate Erectile Dysfunction in Rats. Int J Radiat Oncol Biol Phys 2017; 99:680-688. [PMID: 29280463 DOI: 10.1016/j.ijrobp.2017.06.2449] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/05/2017] [Revised: 06/09/2017] [Accepted: 06/19/2017] [Indexed: 01/22/2023]
Abstract
PURPOSE/OBJECTIVES Radiation-induced erectile-dysfunction (RiED) is one of the most common side effects of radiation therapy (RT) and significantly reduces the quality of life (QoL) of cancer patients. Approximately 50% of prostate cancer patients experience RiED within 3 to 5 years after completion of RT. A series of vascular, muscular, and neurogenic injuries after prostate RT lead to RiED; however, the precise role of RT-induced neurogenic injury in RiED has not been fully established. The cavernous nerves (CN) are postganglionic parasympathetic nerves located beside the prostate gland that assist in penile erection. This study was designed to investigate the role of CN injury, tissue damage, and altered signaling pathways in an RiED rat model. METHODS AND MATERIALS Male rats were exposed to a single dose of 25 Gy prostate-confined RT. Erectile function was evaluated by intracavernous pressure (ICP) measurements conducted both 9 and 14 weeks after RT. Neuronal injury was evaluated in the CN using quantitative polymerase chain reaction, conduction studies, transmission electron microscopy, and immunoblotting. Masson trichrome staining was performed to elucidate fibrosis level in penile tissues. RESULTS There were significant alterations in the ICP (P<.0001) of RT rats versus non-RT rats. TEM analysis showed decreased myelination, increased microvascular damage, and progressive axonal atrophy of the CN fibers after RT. Electrophysiologic analysis showed significant impairment of the CN conduction velocity after RT. RT also significantly increased RhoA/Rho-associated protein kinase 1 (ROCK1) mRNA and protein expression. In addition, penile tissue showed increased apoptosis and fibrosis 14 weeks after RT. CONCLUSIONS RT-induced CN injury may contribute to RiED; this is therefore a rationale for developing novel therapeutic strategies to mitigate CN and tissue damage. Moreover, further investigation of the RhoA/ROCK pathway's role in mitigating RiED is necessary.
Collapse
Affiliation(s)
- Javed Mahmood
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland.
| | - Caroline Q Connors
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Allen A Alexander
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Radmila Pavlovic
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Santanu Samanta
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Sandrine Soman
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Hotaka Matsui
- Department of Urology, University of Tokyo, Tokyo, Japan
| | - Nikolai A Sopko
- James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Trinity J Bivalacqua
- James Buchanan Brady Urological Institute and Department of Urology, The Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Daniel Weinreich
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Cheng-Ying Ho
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - John Eley
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Amit Sawant
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Isabel L Jackson
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Zeljko Vujaskovic
- Division of Translational Radiation Sciences, Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|