1
|
Kim JT, Han SW, Youn DH, Jung H, Lee EH, Kang SM, Cho YJ, Jeon JP. Advanced hydrogel mesh platform with neural stem cells and human umbilical vein endothelial cells for enhanced axonal regeneration. APL Bioeng 2025; 9:026101. [PMID: 40181802 PMCID: PMC11964475 DOI: 10.1063/5.0244057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 03/15/2025] [Indexed: 04/05/2025] Open
Abstract
One of the major obstacles to neural recovery following intracerebral hemorrhage (ICH) is the cavity-like lesion that occurs at the site of the hemorrhage, which impedes axonal regeneration. Here, we aim to address this challenge by investigating the migratory mechanisms of neural stem cells (NSCs) within the cavity in vitro using a hydrogel and endothelial cells. Mouse NSCs (mNSCs) isolated from the subventricular and subgranular zones using the 3D hydrogel culture were evaluated for their neurogenic, extracellular matrix (ECM), and adhesion-related mRNA expression compared to microglia (BV2) and secretory factors of human umbilical vein endothelial cells (HUVECs) in vitro and in vivo conditions. A hydrogel mesh combining mNSCs and HUVECs was developed for its therapeutic potential. mNSCs exhibit high stemness, neurogenesis, and ECM remodeling capabilities. mNSCs demonstrated close interaction with HUVECs and the surrounding vascular structures in in vitro and in vivo studies. Furthermore, mNSCs could degrade high concentrations of fibrin to facilitate migration and adhesion. mNSCs and HUVECs formed mesh networks through cell-cell contacts and maintained the structure through Matrigel support, potentially ensuring sufficient survival and regeneration capabilities. Our proposed hydrogel mesh platform with mNSCs and HUVECs demonstrated successful maintenance of cell survival and provision of structural support for the delivered cells by promoting ECM remodeling and neurogenesis, which may aid in axonal regeneration in the cavity lesions following ICH.
Collapse
Affiliation(s)
- Jong-Tae Kim
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea
| | - Sung Woo Han
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea
| | - Dong Hyuk Youn
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea
| | - Harry Jung
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea
| | - Eun-Ho Lee
- Department of Green Chemical Engineering, Sangmyung University, Cheonan 31066, Republic of Korea
| | - Sung-Min Kang
- Department of Green Chemical Engineering, Sangmyung University, Cheonan 31066, Republic of Korea
| | - Yong-Jun Cho
- Department of Neurosurgery, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea
| | - Jin Pyeong Jeon
- Department of Neurosurgery, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea
| |
Collapse
|
2
|
Deng S, Xie H, Xie B. Cell-based regenerative and rejuvenation strategies for treating neurodegenerative diseases. Stem Cell Res Ther 2025; 16:167. [PMID: 40189500 PMCID: PMC11974143 DOI: 10.1186/s13287-025-04285-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/19/2025] [Indexed: 04/09/2025] Open
Abstract
Neurodegenerative diseases including Alzheimer's and Parkinson's disease are age-related disorders which severely impact quality of life and impose significant societal burdens. Cellular senescence is a critical factor in these disorders, contributing to their onset and progression by promoting permanent cell cycle arrest and reducing cellular function, affecting various types of cells in brain. Recent advancements in regenerative medicine have highlighted "R3" strategies-rejuvenation, regeneration, and replacement-as promising therapeutic approaches for neurodegeneration. This review aims to critically analyze the role of cellular senescence in neurodegenerative diseases and organizes therapeutic approaches within the R3 regenerative medicine paradigm. Specifically, we examine stem cell therapy, direct lineage reprogramming, and partial reprogramming in the context of R3, emphasizing how these interventions mitigate cellular senescence and counteracting aging-related neurodegeneration. Ultimately, this review seeks to provide insights into the complex interplay between cellular senescence and neurodegeneration while highlighting the promise of cell-based regenerative strategies to address these debilitating conditions.
Collapse
Affiliation(s)
- Sixiu Deng
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, 646000, China
- Department of Gastroenterology, The Shapingba Hospital, Chongqing University( People's Hospital of Shapingba District), Chongqing, China
| | - Huangfan Xie
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China.
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, 646000, China.
| | - Bingqing Xie
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China.
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
3
|
Boopathy K, Palaniyandi T, Ravi M, Wahab MRA, Baskar G, Rab SO, Saeed M, Balaramnavar VM. Exploring the potential of stem cell therapy: Applications, types, and future directions. Acta Histochem 2025; 127:152237. [PMID: 40020616 DOI: 10.1016/j.acthis.2025.152237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/14/2025] [Accepted: 02/18/2025] [Indexed: 03/03/2025]
Abstract
One of the most significant treatment approaches now accessible is stem cell therapy. Over the last few decades, a lot of study has been done in this field, and this fascinating feature of plasticity could have therapeutic uses. The potential of stem cells to restore function lost as a result of disease, trauma, congenital defects, and age has made stem cell research a key priority for scientific and medical organizations across the world. Stem cells are a crucial topic of study in regenerative medicine because of their capacity to replace, repair, or regenerate damaged cells, tissues, or organs. As a result, stem cell therapy is being used as a treatment strategy for a number of illnesses. Because stem cells may proliferate indefinitely and generate vast quantities of differentiated cells needed for transplantation, they hold enormous promise for regenerative medicine. Stem cells can be reprogrammed from adult cell types or originate from embryonic or fetal origins. Depending on their availability and place of origin, stem cells can be totipotent, pluripotent, multipotent, oligopotent, or unipotent. With stem cell treatment, many ailments, including diabetes, liver disease, infertility, wounds and traumas, neurological disorders, cardiovascular disease, and cancer, might be cured. Various types of stem cell treatment are described in this review along with their applications in different therapeutic fields, ethical considerations, and advantages and disadvantages.
Collapse
Affiliation(s)
- KeerthiShri Boopathy
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Chennai 600095, India
| | - Thirunavukkarasu Palaniyandi
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Chennai 600095, India; ACS-Advanced Medical Research Institute, Dr. M.G.R Educational and Research Institute, Chennai 600077, India.
| | - Maddaly Ravi
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu 600 116, India
| | | | - Gomathy Baskar
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Chennai 600095, India
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Mohd Saeed
- Department of Biology, College of Science, University of Hail, Hail, Saudi Arabia
| | - Vishal M Balaramnavar
- School of Pharmacy and Research Centre, Sanskriti University, Chhata, Mathura, Uttar Pradesh 281401, India
| |
Collapse
|
4
|
Cha Z, Qiao Y, Lu Q, Wang Q, Lu X, Zhou H, Li T. Research progress and challenges of stem cell therapy for ischemic stroke. Front Cell Dev Biol 2024; 12:1410732. [PMID: 39040041 PMCID: PMC11260720 DOI: 10.3389/fcell.2024.1410732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/17/2024] [Indexed: 07/24/2024] Open
Abstract
Ischemic stroke is a significant global cause of death and disability. Currently, treatment options for acute ischemic stroke are limited to intravenous thrombolysis and mechanical recanalization. Therefore, novel neuroprotective strategies are imperative. Stem cell transplantation possesses the capabilities of differentiation, proliferation, neuronal replacement, nerve pathway reconstruction, secretion of nerve growth factors, and enhancement of the microenvironment; thus, it is a potential therapeutic approach for ischemic stroke. In addition, the immunomodulatory function of stem cells and the combined treatment of stem cells and exosomes exhibit a favorable protective effect on brain injury and neurological dysfunction following stroke. Meanwhile, the theory of microbiota-gut-brain axis provides us with a novel perspective for comprehending and managing neurological diseases. Lastly, stem cell transplantation has demonstrated promising outcomes not only in treating ischemic stroke but also in dealing with other neurological disorders, such as brain tumors. Furthermore, challenges related to the tissue source, delivery method, immune response, and timing of transplantation still need to be addressed to optimize the treatment.
Collapse
Affiliation(s)
- Zaihong Cha
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yisheng Qiao
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Qixiong Lu
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Qiyang Wang
- Department of Orthopedics, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Xiaoyang Lu
- Department of Neurosurgery, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Hu Zhou
- Department of Neurosurgery, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Tao Li
- Research Center for Clinical Medicine, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Institute of Neurosurgery and Neuroscience, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
5
|
Pramanik S, Bala A, Pradhan A. Zebrafish in understanding molecular pathophysiology, disease modeling, and developing effective treatments for Rett syndrome. J Gene Med 2024; 26:e3677. [PMID: 38380785 DOI: 10.1002/jgm.3677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/04/2024] [Accepted: 01/28/2024] [Indexed: 02/22/2024] Open
Abstract
Rett syndrome (RTT) is a rare but dreadful X-linked genetic disease that mainly affects young girls. It is a neurological disease that affects nerve cell development and function, resulting in severe motor and intellectual disabilities. To date, no cure is available for treating this disease. In 90% of the cases, RTT is caused by a mutation in methyl-CpG-binding protein 2 (MECP2), a transcription factor involved in the repression and activation of transcription. MECP2 is known to regulate several target genes and is involved in different physiological functions. Mouse models exhibit a broad range of phenotypes in recapitulating human RTT symptoms; however, understanding the disease mechanisms remains incomplete, and many potential RTT treatments developed in mouse models have not shown translational effectiveness in human trials. Recent data hint that the zebrafish model emulates similar disrupted neurological functions following mutation of the mecp2 gene. This suggests that zebrafish can be used to understand the onset and progression of RTT pathophysiology and develop a possible cure. In this review, we elaborate on the molecular basis of RTT pathophysiology in humans and model organisms, including rodents and zebrafish, focusing on the zebrafish model to understand the molecular pathophysiology and the development of therapeutic strategies for RTT. Finally, we propose a rational treatment strategy, including antisense oligonucleotides, small interfering RNA technology and induced pluripotent stem cell-derived cell therapy.
Collapse
Affiliation(s)
- Subrata Pramanik
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, Assam, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Asis Bala
- Pharmacology and Drug Discovery Research Laboratory, Division of Life Sciences; Institute of Advanced Study in Science and Technology (IASST), An Autonomous Institute Under - Department of Science & Technology (Govt. of India) Vigyan Path, Guwahati, Assam, India
| | - Ajay Pradhan
- Biology, The Life Science Center, School of Science and Technology, Örebro University, Örebro, Sweden
| |
Collapse
|
6
|
Valenti D, Vacca RA. Brain Mitochondrial Bioenergetics in Genetic Neurodevelopmental Disorders: Focus on Down, Rett and Fragile X Syndromes. Int J Mol Sci 2023; 24:12488. [PMID: 37569863 PMCID: PMC10419900 DOI: 10.3390/ijms241512488] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/31/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Abstract
Mitochondria, far beyond their prominent role as cellular powerhouses, are complex cellular organelles active as central metabolic hubs that are capable of integrating and controlling several signaling pathways essential for neurological processes, including neurogenesis and neuroplasticity. On the other hand, mitochondria are themselves regulated from a series of signaling proteins to achieve the best efficiency in producing energy, in establishing a network and in performing their own de novo synthesis or clearance. Dysfunctions in signaling processes that control mitochondrial biogenesis, dynamics and bioenergetics are increasingly associated with impairment in brain development and involved in a wide variety of neurodevelopmental disorders. Here, we review recent evidence proving the emerging role of mitochondria as master regulators of brain bioenergetics, highlighting their control skills in brain neurodevelopment and cognition. We analyze, from a mechanistic point of view, mitochondrial bioenergetic dysfunction as causally interrelated to the origins of typical genetic intellectual disability-related neurodevelopmental disorders, such as Down, Rett and Fragile X syndromes. Finally, we discuss whether mitochondria can become therapeutic targets to improve brain development and function from a holistic perspective.
Collapse
Affiliation(s)
- Daniela Valenti
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Via G. Amendola 122/O, 70126 Bari, Italy
| | - Rosa Anna Vacca
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Via G. Amendola 122/O, 70126 Bari, Italy
| |
Collapse
|
7
|
Fernández Ó, Montalban X, Agüera E, Aladro Y, Alonso A, Arroyo R, Brieva L, Calles C, Costa-Frossard L, Eichau S, García-Domínguez JM, Hernández MÁ, Landete L, Llaneza M, Llufriu S, Meca-Lallana JE, Meca-Lallana V, Mongay-Ochoa N, Moral E, Oreja-Guevara C, Ramió-Torrentà L, Téllez N, Romero-Pinel L, Rodríguez-Antigüedad A. [15th Post-ECTRIMS Meeting: a review of the latest developments presented at the 2022 ECTRIMS Congress (Part I)]. Rev Neurol 2023; 77:19-30. [PMID: 37365721 PMCID: PMC10663806 DOI: 10.33588/rn.7701.2023167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Indexed: 06/28/2023]
Abstract
INTRODUCTION On 4 and 5 November 2022, Madrid hosted the 15th edition of the Post-ECTRIMS Meeting, where neurologists specialised in multiple sclerosis (MS) outlined the most relevant novelties presented at the 2022 ECTRIMS Congress, held in Amsterdam from 26 to 28 October. AIM To synthesise the content presented at the 15th edition of the Post-ECTRIMS Meeting, in an article broken down into two parts. DEVELOPMENT In this first part, the initial events involved in the onset of MS, the role played by lymphocytes and the migration of immune system cells into the central nervous system are presented. It describes emerging biomarkers in body fluids and imaging findings that are predictive of disease progression and useful in the differential diagnosis of MS. It also discusses advances in imaging techniques which, together with a better understanding of the agents involved in demyelination and remyelination processes, provide a basis for dealing with remyelination in the clinical setting. Finally, the mechanisms triggering the inflammatory reaction and neurodegeneration involved in MS pathology are reviewed.
Collapse
Affiliation(s)
- Óscar Fernández
- Hospital Regional Universitario de Málaga. MálagaHospital Regional Universitario de MálagaHospital Regional Universitario de MálagaMálagaSpain
| | - Xavier Montalban
- Hospital Universitari Vall d’Hebron-CEMCATHospital Universitari Vall d’Hebron-CEMCATHospital Universitari Vall d’Hebron-CEMCATBarcelonaSpain
| | - Eduardo Agüera
- Hospital Universitario Reina SofíaHospital Universitario Reina SofíaHospital Universitario Reina SofíaBarcelonaSpain
| | - Yolanda Aladro
- Hospital Universitario de Getafe. Getafe, MadridHospital Universitario de GetafeHospital Universitario de GetafeMadridSpain
| | - Ana Alonso
- Hospital Regional Universitario de Málaga. MálagaHospital Regional Universitario de MálagaHospital Regional Universitario de MálagaMálagaSpain
| | - Rafael Arroyo
- Hospital Universitario QuirónsaludHospital Universitario QuirónsaludHospital Universitario QuirónsaludBarcelonaSpain
| | - Luis Brieva
- Hospital Universitari Arnau de Vilanova- Universitat de Lleida. LleidaHospital Universitari Arnau de Vilanova- Universitat de LleidaHospital Universitari Arnau de Vilanova- Universitat de LleidaLleidaSpain
| | - Carmen Calles
- Hospital Universitario Son Espases. Palma de MallorcaHospital Universitario Son EspasesHospital Universitario Son EspasesPalma de MallorcaSpain
| | - Lucienne Costa-Frossard
- Hospital Universitario Ramón y CajalHospital Universitario Ramón y CajalHospital Universitario Ramón y CajalBarcelonaSpain
| | - Sara Eichau
- Hospital Universitario Virgen Macarena. SevillaHospital Universitario Virgen MacarenaHospital Universitario Virgen MacarenaSevillaSpain
| | - José M. García-Domínguez
- Hospital Universitario Gregorio MarañónHospital Universitario Gregorio MarañónHospital Universitario Gregorio MarañónBarcelonaSpain
| | - Miguel Á. Hernández
- Hospital Nuestra Señora de Candelaria. Santa Cruz de TenerifeHospital Nuestra Señora de CandelariaHospital Nuestra Señora de CandelariaSanta Cruz de TenerifeSpain
| | - Lamberto Landete
- Hospital Universitario Doctor Peset. ValenciaHospital Universitario Doctor PesetHospital Universitario Doctor PesetValenciaSpain
| | - Miguel Llaneza
- Complejo Hospitalario Universitario de Ferrol. El Ferrol, La CoruñaComplejo Hospitalario Universitario de FerrolComplejo Hospitalario Universitario de FerrolEl FerrolSpain
| | - Sara Llufriu
- Hospital Clínic de Barcelona e IDIBAPS. BarcelonaHospital Clínic de Barcelona e IDIBAPSHospital Clínic de Barcelona e IDIBAPSBarcelonaSpain
| | - José E. Meca-Lallana
- Hospital Regional Universitario de Málaga. MálagaHospital Regional Universitario de MálagaHospital Regional Universitario de MálagaMálagaSpain
| | - Virginia Meca-Lallana
- Hospital Clínico Universitario Virgen de la Arrixaca. MurciaHospital Clínico Universitario Virgen de la ArrixacaHospital Clínico Universitario Virgen de la ArrixacaMurciaSpain
| | - Neus Mongay-Ochoa
- Hospital Universitari Vall d’Hebron-CEMCATHospital Universitari Vall d’Hebron-CEMCATHospital Universitari Vall d’Hebron-CEMCATBarcelonaSpain
| | - Ester Moral
- Hospital Sant Joan Despí Moisès Broggi. Sant Joan Despí, BarcelonaHospital Sant Joan Despí Moisès BroggiHospital Sant Joan Despí Moisès BroggiBarcelonaSpain
| | - Celia Oreja-Guevara
- Hospital Clínico San Carlos-IdISSC-UCM. MadridHospital Clínico San Carlos-IdISSC-UCMHospital Clínico San Carlos-IdISSC-UCMMadridSpain
| | - Lluís Ramió-Torrentà
- Departamento de Cièncias Médicas. Universitat de Girona. GironaUniversitat de GironaUniversitat de GironaGironaSpain
| | - Nieves Téllez
- Hospital Clínico Universitario de Valladolid. ValladolidHospital Clínico Universitario de ValladolidHospital Clínico Universitario de ValladolidValladolidSpain
| | - Lucía Romero-Pinel
- Hospital Universitari de Bellvitge- IDIBELL. L’Hospitalet de Llobregat, BarcelonaHospital Universitari de Bellvitge- IDIBELLHospital Universitari de Bellvitge- IDIBELLBarcelonaSpain
| | | |
Collapse
|
8
|
Medina S, Ihrie RA, Irish JM. Learning cell identity in immunology, neuroscience, and cancer. Semin Immunopathol 2023; 45:3-16. [PMID: 36534139 PMCID: PMC9762661 DOI: 10.1007/s00281-022-00976-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 11/19/2022] [Indexed: 12/23/2022]
Abstract
Suspension and imaging cytometry techniques that simultaneously measure hundreds of cellular features are powering a new era of cell biology and transforming our understanding of human tissues and tumors. However, a central challenge remains in learning the identities of unexpected or novel cell types. Cell identification rubrics that could assist trainees, whether human or machine, are not always rigorously defined, vary greatly by field, and differentially rely on cell intrinsic measurements, cell extrinsic tissue measurements, or external contextual information such as clinical outcomes. This challenge is especially acute in the context of tumors, where cells aberrantly express developmental programs that are normally time, location, or cell-type restricted. Well-established fields have contrasting practices for cell identity that have emerged from convention and convenience as much as design. For example, early immunology focused on identifying minimal sets of protein features that mark individual, functionally distinct cells. In neuroscience, features including morphology, development, and anatomical location were typical starting points for defining cell types. Both immunology and neuroscience now aim to link standardized measurements of protein or RNA to informative cell functions such as electrophysiology, connectivity, lineage potential, phospho-protein signaling, cell suppression, and tumor cell killing ability. The expansion of automated, machine-driven methods for learning cell identity has further created an urgent need for a harmonized framework for distinguishing cell identity across fields and technology platforms. Here, we compare practices in the fields of immunology and neuroscience, highlight concepts from each that might work well in the other, and propose ways to implement these ideas to study neural and immune cell interactions in brain tumors and associated model systems.
Collapse
Affiliation(s)
- Stephanie Medina
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rebecca A Ihrie
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Department of Neurological Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jonathan M Irish
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA.
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
9
|
Tian J, Wang H, Zheng S, Ning Y, Zhang X, Niu J, Vladareanu L. sEMG-Based Gain-Tuned Compliance Control for the Lower Limb Rehabilitation Robot during Passive Training. SENSORS (BASEL, SWITZERLAND) 2022; 22:7890. [PMID: 36298256 PMCID: PMC9611623 DOI: 10.3390/s22207890] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/13/2022] [Accepted: 10/14/2022] [Indexed: 06/16/2023]
Abstract
The lower limb rehabilitation robot is a typical man-machine coupling system. Aiming at the problems of insufficient physiological information and unsatisfactory safety performance in the compliance control strategy for the lower limb rehabilitation robot during passive training, this study developed a surface electromyography-based gain-tuned compliance control (EGCC) strategy for the lower limb rehabilitation robot. First, the mapping function relationship between the normalized surface electromyography (sEMG) signal and the gain parameter was established and an overall EGCC strategy proposed. Next, the EGCC strategy without sEMG information was simulated and analyzed. The effects of the impedance control parameters on the position correction amount were studied, and the change rules of the robot end trajectory, man-machine contact force, and position correction amount analyzed in different training modes. Then, the sEMG signal acquisition and feature analysis of target muscle groups under different training modes were carried out. Finally, based on the lower limb rehabilitation robot control system, the influence of normalized sEMG threshold on the robot end trajectory and gain parameters under different training modes was experimentally studied. The simulation and experimental results show that the adoption of the EGCC strategy can significantly enhance the compliance of the robot end-effector by detecting the sEMG signal and improve the safety of the robot in different training modes, indicating the EGCC strategy has good application prospects in the rehabilitation robot field.
Collapse
Affiliation(s)
- Junjie Tian
- Parallel Robot and Mechatronic System Laboratory of Hebei Province, Yanshan University, Qinhuangdao 066004, China
| | - Hongbo Wang
- Parallel Robot and Mechatronic System Laboratory of Hebei Province, Yanshan University, Qinhuangdao 066004, China
- Academy for Engineering & Technology, Fudan University, Shanghai 200433, China
| | - Siyuan Zheng
- Parallel Robot and Mechatronic System Laboratory of Hebei Province, Yanshan University, Qinhuangdao 066004, China
| | - Yuansheng Ning
- Parallel Robot and Mechatronic System Laboratory of Hebei Province, Yanshan University, Qinhuangdao 066004, China
| | - Xingchao Zhang
- Parallel Robot and Mechatronic System Laboratory of Hebei Province, Yanshan University, Qinhuangdao 066004, China
| | - Jianye Niu
- Parallel Robot and Mechatronic System Laboratory of Hebei Province, Yanshan University, Qinhuangdao 066004, China
| | - Luige Vladareanu
- Institute of Solid Mechanics of the Romanian Academy, 010141 Bucharest, Romania
| |
Collapse
|
10
|
Sakowski SA, Chen KS. Stem cell therapy for central nervous system disorders: Metabolic interactions between transplanted cells and local microenvironments. Neurobiol Dis 2022; 173:105842. [PMID: 35988874 PMCID: PMC10117179 DOI: 10.1016/j.nbd.2022.105842] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 10/15/2022] Open
Abstract
Stem cell therapy is a promising and rapidly advancing treatment strategy for a multitude of neurologic disorders. Yet, while early phase clinical trials are being pursued in many disorders, the mechanism of action often remains unclear. One important potential mechanism by which stem cells provide neuroprotection is through metabolic signaling with diseased neurons, glia, and other cell types in the nervous system microenvironment. Early studies exploring such interactions report normalization of glucose metabolism, induction of protective mitochondrial genes, and even interactions with supportive neurovasculature. Local metabolic conditions also impact stem cell biology, which can have a large impact on transplant viability and efficacy. Epigenetic changes that occur in the donor prior to collection of stem cells, and even during in vitro culture conditions, may have effects on stem cell biology that are carried into the host upon stem cell transplantation. Transplanted stem cells also face potentially toxic metabolic microenvironments at the targeted transplant site. Novel approaches for metabolically "preconditioning" stem cells prior to transplant harness metabolic machinery to optimize stem cell survival upon transplant. Ultimately, an improved understanding of the metabolic cross-talk between implanted stem cells and the local nervous system environment, in both disease and injury states, will increase the likelihood of success in translating stem cell therapy to early trials in neurological disease.
Collapse
Affiliation(s)
- Stacey A Sakowski
- Department of Neurology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA.
| | - Kevin S Chen
- Department of Neurology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI 48109, USA; Department of Neurosurgery, University of Michigan, 1500 E. Medical Center Dr, Ann Arbor, MI 48109, USA.
| |
Collapse
|
11
|
Dermitzakis I, Manthou ME, Meditskou S, Miliaras D, Kesidou E, Boziki M, Petratos S, Grigoriadis N, Theotokis P. Developmental Cues and Molecular Drivers in Myelinogenesis: Revisiting Early Life to Re-Evaluate the Integrity of CNS Myelin. Curr Issues Mol Biol 2022; 44:3208-3237. [PMID: 35877446 PMCID: PMC9324160 DOI: 10.3390/cimb44070222] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/14/2022] [Accepted: 07/17/2022] [Indexed: 02/07/2023] Open
Abstract
The mammalian central nervous system (CNS) coordinates its communication through saltatory conduction, facilitated by myelin-forming oligodendrocytes (OLs). Despite the fact that neurogenesis from stem cell niches has caught the majority of attention in recent years, oligodendrogenesis and, more specifically, the molecular underpinnings behind OL-dependent myelinogenesis, remain largely unknown. In this comprehensive review, we determine the developmental cues and molecular drivers which regulate normal myelination both at the prenatal and postnatal periods. We have indexed the individual stages of myelinogenesis sequentially; from the initiation of oligodendrocyte precursor cells, including migration and proliferation, to first contact with the axon that enlists positive and negative regulators for myelination, until the ultimate maintenance of the axon ensheathment and myelin growth. Here, we highlight multiple developmental pathways that are key to successful myelin formation and define the molecular pathways that can potentially be targets for pharmacological interventions in a variety of neurological disorders that exhibit demyelination.
Collapse
Affiliation(s)
- Iasonas Dermitzakis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Maria Eleni Manthou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Soultana Meditskou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Dimosthenis Miliaras
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC 3004, Australia;
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Paschalis Theotokis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
- Correspondence:
| |
Collapse
|
12
|
Uenaka M, Uyeda A, Nakahara T, Muramatsu R. LPA 2 promotes neuronal differentiation and neurite formation in neocortical development. Biochem Biophys Res Commun 2022; 598:89-94. [PMID: 35151977 DOI: 10.1016/j.bbrc.2022.01.109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/27/2022] [Indexed: 11/18/2022]
Abstract
Lysophosphatidic acid (LPA) is a bioactive lipid that activates the G protein-coupled receptors, LPA1-6, which are associated with a wide number of cellular responses including proliferation, migration, differentiation, and survival. Although LPA1-6 are expressed in the developing brain, their functions in brain development are not fully understood. In the present study, we analyzed the temporal expression pattern of LPA receptors (LPARs) during neocortical development and found that LPA2 is highly expressed in neural stem/progenitor cells (NS/PCs) in the embryonic neocortex. LPA2 activation on cultured NS/PCs using GRI977143, a selective LPA2 agonist, promoted neuronal differentiation. LPA2-induced neuronal expansion was inhibited by FR180204, an extracellular signal-regulated kinase 1/2 (Erk1/2) inhibitor, suggesting that LPA2 promotes neuronal differentiation via Erk1/2 signaling. In addition, LPA2 activation promotes neurite elongation and branch formation. These results suggest that LPA2 is a critical regulator of neuronal differentiation and development.
Collapse
Affiliation(s)
- Mizuki Uenaka
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan; Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Akiko Uyeda
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan
| | - Tsutomu Nakahara
- Department of Molecular Pharmacology, Kitasato University School of Pharmaceutical Sciences, 5-9-1 Shirokane, Minato-ku, Tokyo, 108-8641, Japan
| | - Rieko Muramatsu
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-higashi, Kodaira, Tokyo, 187-8502, Japan.
| |
Collapse
|
13
|
Ozgen S, Krigman J, Zhang R, Sun N. Significance of mitochondrial activity in neurogenesis and neurodegenerative diseases. Neural Regen Res 2022; 17:741-747. [PMID: 34472459 PMCID: PMC8530128 DOI: 10.4103/1673-5374.322429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/13/2021] [Accepted: 03/13/2021] [Indexed: 12/11/2022] Open
Abstract
Mitochondria play a multidimensional role in the function and the vitality of the neurological system. From the generation of neural stem cells to the maintenance of neurons and their ultimate demise, mitochondria play a critical role in regulating our neural pathways' homeostasis, a task that is critical to our cognitive health and neurological well-being. Mitochondria provide energy via oxidative phosphorylation for the neurotransmission and generation of an action potential along the neuron's axon. This paper will first review and examine the molecular subtleties of the mitochondria's role in neurogenesis and neuron vitality, as well as outlining the impact of defective mitochondria in neural aging. The authors will then summarize neurodegenerative diseases related to either neurogenesis or homeostatic dysfunction. Because of the significant detriment neurodegenerative diseases have on the quality of life, it is essential to understand their etiology and ongoing molecular mechanics. The mitochondrial role in neurogenesis and neuron vitality is essential. Dissecting and understanding this organelle's role in the genesis and homeostasis of neurons should assist in finding pharmaceutical targets for neurodegenerative diseases.
Collapse
Affiliation(s)
- Serra Ozgen
- Departments of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- College of Medicine, Graduate Research in the Department of Neuroscience, The Ohio State University, Columbus, OH, USA
| | - Judith Krigman
- Departments of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ruohan Zhang
- Departments of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- College of Pharmacy, Department of Graduate Research, The Ohio State University, Columbus, OH, USA
| | - Nuo Sun
- Departments of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
14
|
Kaneko H, Namihira M, Yamamoto S, Numata N, Hyodo K. Oral administration of cyclic glycyl-proline facilitates task learning in a rat stroke model. Behav Brain Res 2022; 417:113561. [PMID: 34509530 DOI: 10.1016/j.bbr.2021.113561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 08/29/2021] [Accepted: 09/01/2021] [Indexed: 11/25/2022]
Abstract
Cyclic glycyl-proline (cGP) exerts neuroprotective effects against ischemic stroke and may promote neural plasticity or network remodeling. We sought to determine to what extent oral administration of cGP could facilitate task learning in rats with ischemic lesions. We trained rats to perform a choice reaction time task using their forepaws. One week after changing the food to pellets containing cGP (no cGP: 0 mg/kg; low cGP: 25 mg/kg; and high cGP: 75 mg/kg), we made a focal ischemic lesion on the left or right forepaw area of the sensorimotor cortex. After recovery of task performance, we altered the correct-response side of the task, and then analyzed the number of training days required for the rat to reach a learning criterion (error rate < 15%) and the regulation of adult neurogenesis in the subventricular zones (SVZs), taking lesion size into account. The low-cGP group required fewer training days for task learning than the no-cGP group. Unexpectedly, rats with larger lesions required fewer training days in the no-cGP and low-cGP groups, but more training days in the high-cGP group. The number of Ki67-immunopositive cells (indicating proliferative cells) in ipsilesional SVZ increased more rapidly in the low-cGP and high-cGP groups than in the no-cGP group. However, lesion size had only a small effect on required training days and the number of Ki67-immunopositive cells. We conclude that oral administration of cGP can facilitate task learning in rats with focal ischemic infarction through neural plasticity and network remodeling, even with minimal neuroprotective effects.
Collapse
Affiliation(s)
- Hidekazu Kaneko
- National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki, Japan.
| | - Masakazu Namihira
- National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki, Japan
| | | | | | - Koji Hyodo
- National Institute of Advanced Industrial Science and Technology (AIST), Ibaraki, Japan
| |
Collapse
|
15
|
Gomez J, Holmes N, Hansen A, Adhikarla V, Gutova M, Rockne RC, Cho H. Mathematical modeling of therapeutic neural stem cell migration in mouse brain with and without brain tumors. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2022; 19:2592-2615. [PMID: 35240798 PMCID: PMC8958926 DOI: 10.3934/mbe.2022119] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Neural stem cells (NSCs) offer a potential solution to treating brain tumors. This is because NSCs can circumvent the blood-brain barrier and migrate to areas of damage in the central nervous system, including tumors, stroke, and wound injuries. However, for successful clinical application of NSC treatment, a sufficient number of viable cells must reach the diseased or damaged area(s) in the brain, and evidence suggests that it may be affected by the paths the NSCs take through the brain, as well as the locations of tumors. To study the NSC migration in brain, we develop a mathematical model of therapeutic NSC migration towards brain tumor, that provides a low cost platform to investigate NSC treatment efficacy. Our model is an extension of the model developed in Rockne et al. (PLoS ONE 13, e0199967, 2018) that considers NSC migration in non-tumor bearing naive mouse brain. Here we modify the model in Rockne et al. in three ways: (i) we consider three-dimensional mouse brain geometry, (ii) we add chemotaxis to model the tumor-tropic nature of NSCs into tumor sites, and (iii) we model stochasticity of migration speed and chemosensitivity. The proposed model is used to study migration patterns of NSCs to sites of tumors for different injection strategies, in particular, intranasal and intracerebral delivery. We observe that intracerebral injection results in more NSCs arriving at the tumor site(s), but the relative fraction of NSCs depends on the location of injection relative to the target site(s). On the other hand, intranasal injection results in fewer NSCs at the tumor site, but yields a more even distribution of NSCs within and around the target tumor site(s).
Collapse
Affiliation(s)
- Justin Gomez
- Department of Mathematics, University of California, Riverside, Riverside, CA 92521, USA
| | - Nathanael Holmes
- Department of Mathematics, University of California, Riverside, Riverside, CA 92521, USA
| | - Austin Hansen
- Department of Mathematics, University of California, Riverside, Riverside, CA 92521, USA
| | - Vikram Adhikarla
- Division of Mathematical Oncology, Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Margarita Gutova
- Department of Stem Cell Biology and Regenerative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Russell C. Rockne
- Division of Mathematical Oncology, Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Heyrim Cho
- Department of Mathematics, University of California, Riverside, Riverside, CA 92521, USA
| |
Collapse
|
16
|
Ricca A, Cascino F, Gritti A. Isolation and Culture of Neural Stem/Progenitor Cells from the Postnatal Periventricular Region. Methods Mol Biol 2022; 2389:11-31. [PMID: 34557998 DOI: 10.1007/978-1-0716-1783-0_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Due to the complexity of the neural stem cell (NSC) niche organization, the lack of specific NSC markers, and the difficulty of long-term tracking these cells and their progeny in vivo, the functional properties of the endogenous NSCs remain largely unexplored. These limitations have led to the development of methodologies to efficiently isolate, expand, and differentiate NSCs ex vivo. We describe here the peculiarities of the neurosphere assay (NSA) as a methodology that allows to efficiently isolate, expand, and differentiate somatic NSCs derived from the postnatal and adult forebrain periventricular region while preserving proliferation, self-renewal, and multipotency, the main attributes that provide their functional identification.
Collapse
Affiliation(s)
- Alessandra Ricca
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federica Cascino
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
17
|
Chen B, An J, Guo YS, Tang J, Zhao JJ, Zhang R, Yang H. Tetramethylpyrazine induces the release of BDNF from BM-MSCs through activation of the PI3K/AKT/CREB pathway. Cell Biol Int 2021; 45:2429-2442. [PMID: 34374467 DOI: 10.1002/cbin.11687] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 08/01/2021] [Accepted: 08/07/2021] [Indexed: 12/27/2022]
Abstract
Compelling evidences suggest that transplantation of bone marrow-derived mesenchymal stem cells (BM-MSCs) can be therapeutically effective for central nervous system (CNS) injuries and neurodegenerative diseases. The therapeutic effect of BM-MSCs mainly attributes to their differentiation into neuron-like cells which replace injured and degenerative neurons. Importantly, the neurotrophic factors released from BM-MSCs can also rescue injured and degenerative neurons, which plays a biologically pivotal role in enhancing neuroregeneration and neurological functional recovery. Tetramethylpyrazine (TMP), the main bioactive ingredient extracted from the traditional Chinese medicinal herb Chuanxiong, has been reported to promote the neuronal differentiation of BM-MSCs. This study aimed to investigate whether TMP regulates the release of neurotrophic factors from BM-MSCs. We examined the effect of TMP on brain-derived neurotrophic factor (BDNF) released from BM-MSCs and elucidated the underlying molecular mechanism. Our results demonstrated that TMP at concentrations of lower than 200 μM increased the release of BDNF in a dose-dependent manner. Furthermore, the effect of TMP on increasing the release of BDNF from BM-MSCs was blocked by inhibiting the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/protein kinase B (AKT)/cAMP-response element binding protein (CREB) pathway. Therefore, we concluded that TMP could induce the release of BDNF from BM-MSCs through activation of the PI3K/AKT/CREB pathway, leading to the formation of neuroprotective and proneurogenic microenvironment. These findings suggest that TMP possesses novel therapeutic potential to promote neuroprotection and neurogenesis through improving the neurotrophic ability of BM-MSCs, which provides a promising nutritional prevention and treatment strategy for CNS injuries and neurodegenerative diseases via the transplantation of TMP-treated BM-MSCs.
Collapse
Affiliation(s)
- Bo Chen
- Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jing An
- Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yun-Shan Guo
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Juan Tang
- Department of Cell Biology, National Translational Science Center for Molecular Medicine, Fourth Military Medical University, Xi'an, China
| | - Jing-Jing Zhao
- Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Rui Zhang
- Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Hao Yang
- Translational Medicine Center, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
18
|
Theotokis P, Kesidou E, Mitsiadou D, Petratos S, Damianidou O, Boziki M, Chatzidimitriou A, Grigoriadis N. Lumbar spine intrathecal transplantation of neural precursor cells promotes oligodendrocyte proliferation in hot spots of chronic demyelination. Brain Pathol 2021; 32:e13040. [PMID: 34845781 PMCID: PMC9245942 DOI: 10.1111/bpa.13040] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 01/02/2023] Open
Abstract
Experimental autoimmune encephalomyelitis (EAE) is a basic and reliable model used to study clinical and pathological hallmarks of multiple sclerosis (MS) in rodents. Several studies suggest neural precursor cells (NPCs) as a significant research tool while reporting that transplanted NPCs are a promising therapeutic approach to treating neurological disorders, such as MS. The main objective was to approach a preclinical, in vivo scenario of oligodendrogenesis with NPCs, targeting the main chronic demyelinated lumbosacral milieu of EAE, via the least invasive delivery method which is lumbar puncture. We utilized MOG35‐55 peptide to induce EAE in C57BL/6 mice and prior to the acute relapse, we intervened with either the traceable GFP+ cellular therapy or saline solution in the intrathecal space of their lumbar spine. A BrdU injection, which enabled us to monitor endogenous proliferation, marked the endpoint 50 days post‐induction (50 dpi). Neuropathology with high‐throughput, triple immunofluorescent, and transmission electron microscopy (TEM) data were extracted and analyzed. The experimental treatment attenuated the chronic phase of EAE (50 dpi; score <1) following an acute, clinical relapse. Myelination and axonal integrity were rescued in the NPC‐treated animals along with suppressed immune populations. The differentiation profile of the exogenous NPCs and endogenous BrdU+ cells was location‐dependent where GFP+‐rich areas drove undifferentiated phenotypes toward the oligodendrocyte lineage. In situ oligodendrocyte enrichment was demonstrated through increased (p < 0.001) gap junction channels of Cx32 and Cx47, reliable markers for proliferative oligodendroglia syncytium. TEM morphometric analysis ultimately manifested an increased g‐ratio in lumbosacral fibers of the recovered animals (p < 0.001). Herein, we suggest that a single, lumbar intrathecal administration of NPCs capacitated a viable cellular load and resulted in clinical and pathological amelioration, stimulating resident OPCs to overcome the remyelination failure in EAE demyelinating locale.
Collapse
Affiliation(s)
- Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece
| | - Dimitra Mitsiadou
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, Victoria, Australia
| | - Olympia Damianidou
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece
| | - Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece
| | | | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece.,Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece
| |
Collapse
|
19
|
Adult Human Multipotent Neural Cells Could Be Distinguished from Other Cell Types by Proangiogenic Paracrine Effects via MCP-1 and GRO. Stem Cells Int 2021; 2021:6737288. [PMID: 34434240 PMCID: PMC8380502 DOI: 10.1155/2021/6737288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/28/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022] Open
Abstract
Adult human multipotent neural cells (ahMNCs) are unique cells derived from adult human temporal lobes. They show multipotent differentiation potentials into neurons and astrocytes. In addition, they possess proangiogenic capacities. The objective of this study was to characterize ahMNCs in terms of expression of cell type-specific markers, in vitro differentiation potentials, and paracrine factors compared with several other cell types including fetal neural stem cells (fNSCs) to provide detailed molecular and functional features of ahMNCs. Interestingly, the expression of cell type-specific markers of ahMNCs could not be differentiated from those of pericytes, mesenchymal stem cells (MSCs), or fNSCs. In contrast, differentiation potentials of ahMNCs and fNSCs into neural cells were higher than those of other cell types. Compared with MSCs, ahMNCs showed lower differentiation capacities into osteogenic and adipogenic cells. Moreover, ahMNCs uniquely expressed higher levels of MCP-1 and GRO family paracrine factors than fNSCs and MSCs. These high levels of MCP-1 and GRO family mediated in vivo proangiogenic effects of ahMNCs. These results indicate that ahMNCs have their own distinct characteristics that could distinguish ahMNCs from other cell types. Characteristics of ahMNCs could be utilized further in the preclinical and clinical development of ahMNCs for regenerative medicine. They could also be used as experimental references for other cell types including fNSCs.
Collapse
|
20
|
Wang D, Wang K, Liu Z, Wang Z, Wu H. Valproic Acid Labeled Chitosan Nanoparticles Promote the Proliferation and Differentiation of Neural Stem Cells After Spinal Cord Injury. Neurotox Res 2021; 39:456-466. [PMID: 33247828 DOI: 10.1007/s12640-020-00304-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 10/05/2020] [Accepted: 10/21/2020] [Indexed: 12/12/2022]
Abstract
Chitosan nanoparticles and valproic acid are demonstrated as the protective agents in the treatment of spinal cord injury (SCI). However, the effects of valproic acid-labeled chitosan nanoparticles (VA-CN) on endogenous spinal cord neural stem cells (NSCs) following SCI and the underlying mechanisms involved remain to be elucidated. In this study, the VA-CN was constructed and the effects of VA-CN on NSCs were assessed in a rat model of SCI. We found VA-CN treatment promoted recovery of the tissue and locomotive function following SCI. Moreover, administration of VA-CN significantly enhanced neural stem cell proliferation and the expression levels of neurotrophic factors following SCI. Furthermore, administration of VA-CN led to a decrease in the number of microglia following SCI. In addition, VA-CN treatment significantly increased the Tuj 1- positive cells in the spinal cord of the SCI rats, suggesting that VA-CN could enhance the differentiation of NSCs following SCI. In conclusion, these results demonstrated that VA-CN could improve the functional and histological recovery through promoting the proliferation and differentiation of NSCs following SCI, which would provide a newly potential therapeutic manner for the treatment of SCI.
Collapse
Affiliation(s)
- Dimin Wang
- School of Medicine, Zhejiang University, Hangzhou, China
- College of Basic Medical Sciences, Second Military Medical University, Shanghai, China
| | - Kai Wang
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Zhenlei Liu
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Zonglin Wang
- College of Basic Medical Sciences, Second Military Medical University, Shanghai, China
| | - Hao Wu
- Department of Neurosurgery, Xuanwu Hospital of Capital Medical University, Beijing, China.
| |
Collapse
|
21
|
Yang Y, Fan Y, Zhang H, Zhang Q, Zhao Y, Xiao Z, Liu W, Chen B, Gao L, Sun Z, Xue X, Shu M, Dai J. Small molecules combined with collagen hydrogel direct neurogenesis and migration of neural stem cells after spinal cord injury. Biomaterials 2020; 269:120479. [PMID: 33223332 DOI: 10.1016/j.biomaterials.2020.120479] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 09/28/2020] [Accepted: 10/18/2020] [Indexed: 12/22/2022]
Abstract
Complete spinal cord injury (SCI) leads to cell death, interruption of axonal connections and permanent functional impairments. In the development of SCI treatments, cell transplantation combined with biomaterial-growth factor-based therapies have been widely studied. Another avenue worth exploring is the generation of neurons from endogenous neural stem cells (NSCs) or reactive astrocytes activated by SCI. Here, we screened a combination of four small molecules, LDN193189, SB431542, CHIR99021 and P7C3-A20, that can increase neuronal differentiation of mouse and rat spinal cord NSCs. Moreover, the small molecules loaded in an injectable collagen hydrogel induced neurogenesis and inhibited astrogliogenesis of endogenous NSCs in the injury site, which usually differentiate into astrocytes under pathological conditions. Meanwhile, induced neurons migrated into the non-neural lesion core, and genetic fate mapping showed that neurons mainly originated from NSCs in the parenchyma, but not from the central canal of the spinal cord. The neuronal regeneration in the lesion sites resulted in some recovery of locomotion. Our findings indicate that the combined treatment of small molecules and collagen hydrogel is a potential therapeutic strategy for SCI by inducing in situ endogenous NSCs to form neurons and restore damaged functions.
Collapse
Affiliation(s)
- Yaming Yang
- 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
| | - Yongheng Fan
- 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
| | - Haipeng Zhang
- 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
| | - Qi Zhang
- 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
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wenbin Liu
- 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
| | - Lin Gao
- 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
| | - Zheng 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
| | - Xiaoyu Xue
- 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
| | - Muya Shu
- 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
| | - 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.
| |
Collapse
|
22
|
In Vitro Oxygen-Glucose Deprivation-Induced Stroke Models with Human Neuroblastoma Cell- and Induced Pluripotent Stem Cell-Derived Neurons. Stem Cells Int 2020; 2020:8841026. [PMID: 33178286 PMCID: PMC7647751 DOI: 10.1155/2020/8841026] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/07/2020] [Accepted: 10/07/2020] [Indexed: 12/20/2022] Open
Abstract
Stroke is a devastating neurological disorder and one of the leading causes of mortality and disability. To understand the cellular and molecular mechanisms of stroke and to develop novel therapeutic approaches, two different in vitro human cell-based stroke models were established using oxygen-glucose deprivation (OGD) conditions. In addition, the effect of adipose stem cells (ASCs) on OGD-induced injury was studied. In the present study, SH-SY5Y human neuroblastoma cells and human induced pluripotent stem cells (hiPSCs) were differentiated into neurons, cultured under OGD conditions (1% O2) for 24 h, and subjected to a reperfusion period for 24 or 72 h. After OGD, ASCs were cocultured with neurons on inserts for 24 or 72 h to study the neuroprotective potential of ASCs. The effect of OGD and ASC coculture on the viability, apoptosis, and proliferation of and axonal damage to neuronal cells was studied. The results showed that OGD conditions induced cytotoxicity and apoptosis of SH-SY5Y- and hiPSC-derived neurons, although more severe damage was detected in SH-SY5Y-derived neurons than in hiPSC-derived neurons. Coculture with ASCs was protective for neurons, as the number of dead ASC-cocultured neurons was lower than that of control cells, and coculture increased the proliferation of both cell types. To conclude, we developed in vitro human cell-based stroke models in SH-SY5Y- and hiPSC-derived neurons. This was the first time hiPSCs were used to model stroke in vitro. Since OGD had different effects on the studied cell types, this study highlights the importance of using several cell types in in vitro studies to confirm the outcomes of the study. Here, ASCs exerted a neuroprotective effect by increasing the proliferation and decreasing the death of SH-SY5Y- and hiPSC-derived neurons after OGD.
Collapse
|
23
|
Sharma A, Gokulchandran N, Kulkarni P, Mullangi SK, Bhagawanani K, Ganar V, Sane H, Badhe P. Multiple Cellular Therapies Along with Neurorehabilitation in Spastic Diplegic Cerebral Palsy: A Case Report. INNOVATIONS IN CLINICAL NEUROSCIENCE 2020; 17:31-34. [PMID: 33898099 PMCID: PMC7819579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Cerebral palsy (CP) is a chronic childhood disorder that is characterized by a group of motor and cognitive impairments, resulting in abnormal movement patterns, loss of motor control, incoordination, and unbalanced posture. It can also have an impact on fine motor skills, gross motor skills, and oral motor functioning. Currently, the treatment of CP is palliative and does not cure the disease pathology. Hence, there is a need for an intervention that might be able to alter the core pathology. Autologous bone marrow mononuclear cells (BMMNC) transplantation is one of the novel treatment strategies in recent years. In this study, we presented the case of a 4-year-old male child with spastic diplegic CP who underwent two intrathecal transplantations at interval of seven months with autologous BMMNC along with neurorehabilitation program. During an overall 16-month follow-up, significant improvements were observed in motor control, coordination, balance, sitting tolerance, and memory. The abnormal 'W' sitting posture and scissoring gait pattern of the patient resolved. Started sitting with good head, trunk, and pelvic alignment and attained regular gait pattern; the patient started to walk independently without support as well. On objective scale, Gross Motor Functional Measure score improved from 60.67 to 81.78. The patient's Gross Motor Functional Classification System grade improved from Level 3 to Level 2, and Functional Independent Measure score improved from 97 to 99. A comparative positron emission tomography-computed tomography (PET CT) brain scan was performed before and seven months after the first intervention, which revealed improvement in the metabolism of the anterior cingulate lobe, parietal cortex, medial temporal cortex, thalamus, basal ganglia, and cerebellum. No adverse events were recorded throughout the study. Thus, multiple cellular therapies, along with neurorehabilitation, might be a novel safe, feasible option to enhance recovery in CP.
Collapse
Affiliation(s)
- Alok Sharma
- Drs. Sharma and Gokulchandran are with the Department of Medical Services and Clinical Research at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Ms. Kulkani and Drs. Mullangi and Sane are with the Department of Research and Development at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Dr. Bhagawanani and Mr. Ganar are with the Department of Neurorehabilitation at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Dr. Badhe is with the Department of Regenerative Laboratory Services at NeuroGen Brain and Spine Institute in Navi Mumbai, India
| | - Nandini Gokulchandran
- Drs. Sharma and Gokulchandran are with the Department of Medical Services and Clinical Research at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Ms. Kulkani and Drs. Mullangi and Sane are with the Department of Research and Development at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Dr. Bhagawanani and Mr. Ganar are with the Department of Neurorehabilitation at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Dr. Badhe is with the Department of Regenerative Laboratory Services at NeuroGen Brain and Spine Institute in Navi Mumbai, India
| | - Pooja Kulkarni
- Drs. Sharma and Gokulchandran are with the Department of Medical Services and Clinical Research at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Ms. Kulkani and Drs. Mullangi and Sane are with the Department of Research and Development at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Dr. Bhagawanani and Mr. Ganar are with the Department of Neurorehabilitation at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Dr. Badhe is with the Department of Regenerative Laboratory Services at NeuroGen Brain and Spine Institute in Navi Mumbai, India
| | - Sandhya Kiran Mullangi
- Drs. Sharma and Gokulchandran are with the Department of Medical Services and Clinical Research at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Ms. Kulkani and Drs. Mullangi and Sane are with the Department of Research and Development at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Dr. Bhagawanani and Mr. Ganar are with the Department of Neurorehabilitation at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Dr. Badhe is with the Department of Regenerative Laboratory Services at NeuroGen Brain and Spine Institute in Navi Mumbai, India
| | - Khushboo Bhagawanani
- Drs. Sharma and Gokulchandran are with the Department of Medical Services and Clinical Research at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Ms. Kulkani and Drs. Mullangi and Sane are with the Department of Research and Development at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Dr. Bhagawanani and Mr. Ganar are with the Department of Neurorehabilitation at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Dr. Badhe is with the Department of Regenerative Laboratory Services at NeuroGen Brain and Spine Institute in Navi Mumbai, India
| | - Vishal Ganar
- Drs. Sharma and Gokulchandran are with the Department of Medical Services and Clinical Research at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Ms. Kulkani and Drs. Mullangi and Sane are with the Department of Research and Development at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Dr. Bhagawanani and Mr. Ganar are with the Department of Neurorehabilitation at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Dr. Badhe is with the Department of Regenerative Laboratory Services at NeuroGen Brain and Spine Institute in Navi Mumbai, India
| | - Hemangi Sane
- Drs. Sharma and Gokulchandran are with the Department of Medical Services and Clinical Research at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Ms. Kulkani and Drs. Mullangi and Sane are with the Department of Research and Development at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Dr. Bhagawanani and Mr. Ganar are with the Department of Neurorehabilitation at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Dr. Badhe is with the Department of Regenerative Laboratory Services at NeuroGen Brain and Spine Institute in Navi Mumbai, India
| | - Prerna Badhe
- Drs. Sharma and Gokulchandran are with the Department of Medical Services and Clinical Research at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Ms. Kulkani and Drs. Mullangi and Sane are with the Department of Research and Development at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Dr. Bhagawanani and Mr. Ganar are with the Department of Neurorehabilitation at NeuroGen Brain and Spine Institute in Navi Mumbai, India
- Dr. Badhe is with the Department of Regenerative Laboratory Services at NeuroGen Brain and Spine Institute in Navi Mumbai, India
| |
Collapse
|
24
|
Fernandes MB, Costa M, Ribeiro MF, Siquenique S, Sá Santos S, Martins J, Coelho AV, Silva MFB, Rodrigues CMP, Solá S. Reprogramming of Lipid Metabolism as a New Driving Force Behind Tauroursodeoxycholic Acid-Induced Neural Stem Cell Proliferation. Front Cell Dev Biol 2020; 8:335. [PMID: 32582686 PMCID: PMC7286385 DOI: 10.3389/fcell.2020.00335] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022] Open
Abstract
Recent evidence suggests that neural stem cell (NSC) fate is highly dependent on mitochondrial bioenergetics. Tauroursodeoxycholic acid (TUDCA), an endogenous neuroprotective bile acid and a metabolic regulator, stimulates NSC proliferation and enhances adult NSC pool in vitro and in vivo. In this study, we dissected the mechanism triggered by this proliferation-inducing molecule, namely in mediating metabolic reprogramming. Liquid chromatography coupled with mass spectrometry (LC-MS) based detection of differential proteomics revealed that TUDCA reduces the mitochondrial levels of the long-chain acyl-CoA dehydrogenase (LCAD), an enzyme crucial for β-oxidation of long-chain fatty acids (FA). TUDCA impact on NSC mitochondrial proteome was further confirmed, including in neurogenic regions of adult rats. We show that LCAD raises throughout NSC differentiation, while its silencing promotes NSC proliferation. In contrast, nuclear levels of sterol regulatory element-binding protein (SREBP-1), a major transcription factor of lipid biosynthesis, changes in the opposite manner of LCAD, being upregulated by TUDCA. In addition, alterations in some metabolic intermediates, such as palmitic acid, also supported the TUDCA-induced de novo lipogenesis. More interestingly, a metabolic shift from FA to glucose catabolism appears to occur in TUDCA-treated NSCs, since mitochondrial levels of pyruvate dehydrogenase E1-α (PDHE1-α) were significant enhanced by TUDCA. At last, the mitochondria-nucleus translocation of PDHE1-α was potentiated by TUDCA, associated with an increase of H3-histones and acetylated forms. In conclusion, TUDCA-induced proliferation of NSCs involves metabolic plasticity and mitochondria-nucleus crosstalk, in which nuclear PDHE1-α might be required to assure pyruvate-derived acetyl-CoA for histone acetylation and NSC cycle progression.
Collapse
Affiliation(s)
- Marta B Fernandes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Márcia Costa
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Filipe Ribeiro
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Sónia Siquenique
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Sónia Sá Santos
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Joana Martins
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Ana V Coelho
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Margarida F B Silva
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Cecília M P Rodrigues
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Susana Solá
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
25
|
Villoslada P, Steinman L. New targets and therapeutics for neuroprotection, remyelination and repair in multiple sclerosis. Expert Opin Investig Drugs 2020; 29:443-459. [DOI: 10.1080/13543784.2020.1757647] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Pablo Villoslada
- Department of Psychiatry and Behavioural Sciences & Department of Neurology and Neurological Sciences, Stanford University, California, CA, USA
| | - Lawrence Steinman
- Department of Psychiatry and Behavioural Sciences & Department of Neurology and Neurological Sciences, Stanford University, California, CA, USA
| |
Collapse
|
26
|
Denninger JK, Chen X, Turkoglu AM, Sarchet P, Volk AR, Rieskamp JD, Yan P, Kirby ED. Defining the adult hippocampal neural stem cell secretome: In vivo versus in vitro transcriptomic differences and their correlation to secreted protein levels. Brain Res 2020; 1735:146717. [PMID: 32035887 DOI: 10.1016/j.brainres.2020.146717] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 02/03/2020] [Accepted: 02/05/2020] [Indexed: 01/08/2023]
Abstract
Adult hippocampal neural stem and progenitor cells (NSPCs) secrete a variety of proteins that affect tissue function. Though several individual NSPC-derived proteins have been shown to impact key cellular processes, a broad characterization is lacking. Secretome profiling of low abundance stem cell populations is typically achieved via proteomic characterization of in vitro, isolated cells. Here, we identified hundreds of secreted proteins in conditioned media from in vitro adult mouse hippocampal NSPCs using an antibody array and mass spectrometry. Comparison of protein abundance between antibody array and mass spectrometry plus quantification of several key secreted proteins by ELISA revealed notable disconnect between methods in what proteins were identified as being high versus low abundance, suggesting that data from antibody arrays in particular should be approached with caution. We next assessed the NSPC secretome on a transcriptional level with single cell and bulk RNA sequencing (RNAseq) of cultured NSPCs. Comparison of RNAseq transcript levels of highly secreted proteins revealed that quantification of gene expression did not necessarily predict relative protein abundance. Interestingly, comparing our in vitro NSPC gene expression data with similar data from freshly isolated, in vivo hippocampal NSPCs revealed strong correlations in global gene expression between in vitro and in vivo NSPCs. Understanding the components and functions of the NSPC secretome is essential to understanding how these cells may modulate the hippocampal neurogenic niche. Cumulatively, our data emphasize the importance of using proteomics in conjunction with transcriptomics and highlights the need for better methods of unbiased secretome profiling.
Collapse
Affiliation(s)
- Jiyeon K Denninger
- Department of Psychology, College of Arts and Sciences, The Ohio State University, United States
| | - Xi Chen
- Comprehensive Cancer Center, The Ohio State University, United States
| | - Altan M Turkoglu
- College of Arts and Sciences, The Ohio State University, United States
| | - Patricia Sarchet
- Comprehensive Cancer Center, The Ohio State University, United States
| | - Abby R Volk
- College of Arts and Sciences, The Ohio State University, United States
| | - Joshua D Rieskamp
- Neuroscience Graduate Program, The Ohio State University, United States
| | - Pearlly Yan
- Comprehensive Cancer Center, The Ohio State University, United States; Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, United States
| | - Elizabeth D Kirby
- Department of Psychology, College of Arts and Sciences, The Ohio State University, United States; Department of Neuroscience, The Ohio State University, United States; Chronic Brain Injury Initiative, The Ohio State University, United States.
| |
Collapse
|
27
|
Haldar S, Ghosh S, Kumar V, Roy P, Lahiri D. The Evolving Neural Tissue Engineering Landscape of India. ACS APPLIED BIO MATERIALS 2019; 2:5446-5459. [PMID: 35021543 DOI: 10.1021/acsabm.9b00567] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The healthcare sector in India is witnessing unprecedented advancement. Tissue engineering has become an integral part of healthcare and medicine, particularly where treatments involve functional restoration of any injured or deceased part of the body. Not falling behind much with the progressing medical and healthcare sector of India, tissue engineering is also gaining momentum in the country. Out of several arenas of tissue engineering, India has made its mark in orthopedic and bone regeneration, cosmetic and skin regeneration, and very importantly neural regeneration. There are several articles reviewing the progress and prospects of orthopedic and skin regeneration research in India. However, there is no systematic review on progress, prospects, and pitfalls associated with neural tissue engineering in Indian context. The existing ones mainly focus on the technical advancements in the field from a global perspective. Therefore, it is worthwhile to have an organized look at the evolving neural tissue engineering landscape of India. This review will walk the readers systematically through different aspects of the topic. The review starts with an introduction to the nervous system to help readers appreciate the complexity that must be dealt with while engineering neural tissue. This is followed with a global picture of the neural tissue engineering, prominent research groups working on neural tissue engineering in India, factors that have and are currently molding the prospects of this field, and concluding with an overall perspective on present and future of neural tissue engineering in India.
Collapse
|
28
|
Overexpression of Shrm4 promotes proliferation and differentiation of neural stem cells through activation of GABA signaling pathway. Mol Cell Biochem 2019; 463:115-126. [PMID: 31654185 DOI: 10.1007/s11010-019-03634-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 09/25/2019] [Indexed: 10/25/2022]
Abstract
Shrm4 is a protein that is exclusively expressed in polarized tissues. The physiological function of Shrm4 in the brain was required to be elucidated. Thus, we aimed to explore how the Shrm4-mediated gamma-aminobutyric acid (GABA) pathway affected neural stem cells (NSCs). At first, the Nestin expression in cultured NSCs was identified. After determination of the interaction of Shrm4 and GABAB1, a series of in vitro experiment were performed to detect cell proliferation, the ability of cell colony formation, degree that NSCs differentiated into neurons, the apoptosis rate, and cell cycle. The levels of Shrm4, GABAB1, Bcl-2-associated protein x (Bax), B cell lymphoma 2 (Bcl-2), cleaved Caspase-3, microtubule-associated protein 2 (MAP-2) as well as suppressor of cytokine signaling 2 (SOCS2) were detected to further assess the role of Shrm4 and GABA pathway in NSCs. Initially, we found that Shrm4 could bind to GABAB1, and overexpression of Shrm4 or activation of GABAB1 increased the number of positive cells, and promoted cell viability, colony formation rate and differentiation of NSCs. After overexpression of Shrm4 or activation of GABAB1, cells in the G1 phase were decreased, while those in the S phase were increased with an inhibited cell apoptosis rate in the NSCs. Besides, the overexpression of Shrm4 or activation of GABAB1 upregulated the levels of Shrm4, GABAB1, Bcl-2, MAP-2 and SOCS2, while downregulated Bax and cleaved Caspase-3 in NSCs. Overall, overexpression of Shrm4 activated GABAB1 to stimulate the proliferation and differentiation of NSCs. Thus, Shrm4 might be considered as a novel target for promoting the proliferation and differentiation of NSCs.
Collapse
|
29
|
Watanabe TK. A Review of Stem Cell Therapy for Acquired Brain Injuries and Neurodegenerative Central Nervous System Diseases. PM R 2019; 10:S151-S156. [PMID: 30269801 DOI: 10.1016/j.pmrj.2018.07.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Revised: 06/25/2018] [Accepted: 07/02/2018] [Indexed: 12/13/2022]
Abstract
Cell-based therapies have been the subject of much discussion regarding their potential role in enhancing central nervous system function for a number of pathologic conditions. Much of the current research has been in preclinical trials, with clinical trials in the phase I or I/II stage. Nevertheless, there is considerable interest in the public about the potential regenerative role that stem cells may have in improving function for these neurologic conditions. This review will describe the different types of stem cells that are available, review their possible effects, and discuss some of the variables that investigators need to consider when designing their studies. Current clinical research in the areas of stroke, traumatic brain injury, and neurodegenerative diseases (amyotrophic lateral sclerosis and Parkinson disease) will be reviewed. As this article is aimed at a rehabilitation audience, outcome measures, and the role of concurrent rehabilitation therapies will also be mentioned.
Collapse
Affiliation(s)
- Thomas K Watanabe
- MossRehab at Elkins Park / Einstein Healthcare Network, 60 Township Line Road, Elkins Park, PA 19027(∗).
| |
Collapse
|
30
|
Functions of subventricular zone neural precursor cells in stroke recovery. Behav Brain Res 2019; 376:112209. [PMID: 31493429 DOI: 10.1016/j.bbr.2019.112209] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/11/2019] [Accepted: 09/03/2019] [Indexed: 12/16/2022]
Abstract
The proliferation and ectopic migration of neural precursor cells (NPCs) in response to ischemic brain injury was first reported two decades ago. Since then, studies of brain injury-induced subventricular zone cytogenesis, primarily in rodent models, have provided insight into the cellular and molecular determinants of this phenomenon and its modulation by various factors. However, despite considerable correlational evidence-and some direct evidence-to support contributions of NPCs to behavioral recovery after stroke, the causal mechanisms have not been identified. Here we discuss the subventricular zone cytogenic response and its possible roles in brain injury and disease, focusing on rodent models of stroke. Emerging evidence suggests that NPCs can modulate harmful responses and enhance reparative responses to neurologic diseases. We speculatively identify four broad functions of NPCs in the context of stroke: cell replacement, cytoprotection, remodeling of residual tissue, and immunomodulation. Thus, NPCs may have pleiotropic functions in supporting behavioral recovery after stroke.
Collapse
|
31
|
Adult Neurogenesis in the Subventricular Zone and Its Regulation After Ischemic Stroke: Implications for Therapeutic Approaches. Transl Stroke Res 2019; 11:60-79. [DOI: 10.1007/s12975-019-00717-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 06/13/2019] [Accepted: 06/27/2019] [Indexed: 12/21/2022]
|
32
|
Stratton JA, Shah P, Sinha S, Crowther E, Biernaskie J. A tale of two cousins: Ependymal cells, quiescent neural stem cells and potential mechanisms driving their functional divergence. FEBS J 2019; 286:3110-3116. [PMID: 31111999 DOI: 10.1111/febs.14930] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/20/2019] [Accepted: 05/15/2019] [Indexed: 12/18/2022]
Abstract
Recent work has suggested that stem cells exhibit far greater heterogeneity than initially thought. Indeed, their dynamic nature and shared traits with surrounding niche cells have made prospective identification of adult neural stem cells (NSCs) challenging. Refined fate mapping strategies and single-cell omics techniques have begun to clarify functionally distinct states within the adult NSC pool, the molecular signatures that govern these states, and the functional contributions/interactions with neighboring cells within the subventricular niche. Ependymal cells are the epithelial cells which line the ventricular system and reside in the same niche as NSCs. Our own work has revealed that, despite sharing similar embryonic origins with NSCs and close geographic proximity, ependymal cells are transcriptionally distinct and fail to exhibit stem cell function in vivo, even following injury. Intriguingly, comparison of ependymal cells with qNSCs revealed transcriptional signatures that are largely overlapping, suggesting that post-transcriptional regulation might underlie their divergent phenotypes. Additional analysis of ependymal versus qNSC gene regulatory network activation supports this notion. This Viewpoint summarizes the historical confusion regarding the identity of NSCs within the lateral ventricle niche and describes recent work that provides greater appreciation for the diverse functional states within the NSC niche.
Collapse
Affiliation(s)
- Jo Anne Stratton
- Hotchkiss Brain Institute, University of Calgary, Canada.,Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Canada
| | - Prajay Shah
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Canada
| | - Sarthak Sinha
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Canada
| | - Emilie Crowther
- Hotchkiss Brain Institute, University of Calgary, Canada.,Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Canada
| | - Jeff Biernaskie
- Hotchkiss Brain Institute, University of Calgary, Canada.,Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Canada
| |
Collapse
|
33
|
miR-26a prevents neural stem cells from apoptosis via β-catenin signaling pathway in cardiac arrest-induced brain damage. Biosci Rep 2019; 39:BSR20181635. [PMID: 30992390 PMCID: PMC6522814 DOI: 10.1042/bsr20181635] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 03/27/2019] [Accepted: 04/12/2019] [Indexed: 12/28/2022] Open
Abstract
Neural stem cells (NSCs) transplantation is one of the most promising strategies for the treatment of CA-induced brain damage. The transplanted NSCs could differentiate into new neuron and replace the damaged one. However, the poor survival of NSCs in severe hypoxic condition is the limiting step to make the best use of this kind of therapy. In the present study, we investigated whether the overexpression of miR-26a improves the survival of NSCs in hypoxic environment in vitro and in vivo. In vitro hypoxia injury model is established in NSCs by CoCl2 treatment, and in vivo cardiac arrest (CA) model is established in Sprague-Dawley (SD) rats. Quantitative real-time polymerase chain reaction is used to detect the mRNA level and Western blot is used to examine the protein level of indicated genes. TUNEL staining and flow cytometry are applied to evaluate apoptosis. Dual-luciferase reporter assay is utilized to analyze the target gene of miR-26a. The expression of miR-26a is reduced in both in vitro and in vivo hypoxic model. MiR-26a directly targets 3′-UTR of glycogen synthase kinase 3β (GSK-3β), resulting in increased β-catenin expression and decreased apoptosis of NSCs. Overexpression of miR-26a in transplanted NSCs improves the survival of NSCs and neurological function in CA rats. MiR-26a prevents NSCs from apoptosis by activating β-catenin signaling pathway in CA-induced brain damage model. Modulating miR-26a expression could be a potential strategy to attenuate brain damage induced by CA.
Collapse
|
34
|
Zhou H, Wei M, Lu L, Chu T, Li X, Fu Z, Liu J, Kang Y, Liu L, Lou Y, Zhang C, Gao Y, Kong X, Feng S. Angiopoietin-2 induces the neuronal differentiation of mouse embryonic NSCs via phosphatidylinositol 3 kinase-Akt pathway-mediated phosphorylation of mTOR. Am J Transl Res 2019; 11:1895-1907. [PMID: 30972213 PMCID: PMC6456538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 02/13/2019] [Indexed: 06/09/2023]
Abstract
The fate of neural stem cells (NSCs) is decided by numerous growth factors. Among these factors, the well-known angiogenic factor angiopoietin-2 (Ang-2) has been revealed to participate in neurogenesis separate from its role in angiogenesis. However, the effect of Ang-2 on the fate determination of mouse embryonic NSCs and the underlying mechanism remain unclear. This result of this study indicated that treatment of mouse embryonic NSCs with 200 ng/ml Ang-2 significantly promoted neuronal differentiation without affecting glial differentiation, and mammalian target of rapamycin (mTOR) was phosphorylated in a phosphatidylinositol 3-kinase (PI3K)/Akt-dependent manner during this process. Rapamycin, a specific mTOR inhibitor, suppressed the increase in neuronal differentiation stimulated by Ang-2, and this suppression did not result from an effect of Ang-2 or rapamycin on the apoptosis of differentiated NSCs. Collectively, our research demonstrates that PI3K/Akt pathway-mediated mTOR phosphorylation plays an important role in the Ang-2-enhanced neuronal differentiation of mouse embryonic NSCs.
Collapse
Affiliation(s)
- Hengxing Zhou
- Department of Orthopedics, Tianjin Medical University General HospitalNo. 154 Anshan Road, Heping District, Tianjin 300052, PR China
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in The Central Nervous System, Ministry of EducationTianjin City, No. 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Meng Wei
- Key Laboratory of Immuno Microenvironment and Disease of The Educational Ministry of China, Department of Immunology, Tianjin Medical UniversityNo. 22 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Lu Lu
- Department of Orthopedics, Tianjin Medical University General HospitalNo. 154 Anshan Road, Heping District, Tianjin 300052, PR China
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in The Central Nervous System, Ministry of EducationTianjin City, No. 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Tianci Chu
- Kosair Children’s Hospital Research Institute at The Department of Pediatrics, University of Louisville School of MedicineLouisville, Kentucky 40202, USA
| | - Xueying Li
- Key Laboratory of Immuno Microenvironment and Disease of The Educational Ministry of China, Department of Immunology, Tianjin Medical UniversityNo. 22 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Zheng Fu
- Key Laboratory of Immuno Microenvironment and Disease of The Educational Ministry of China, Department of Immunology, Tianjin Medical UniversityNo. 22 Qixiangtai Road, Heping District, Tianjin 300070, PR China
| | - Jun Liu
- Department of Orthopedics, Tianjin Medical University General HospitalNo. 154 Anshan Road, Heping District, Tianjin 300052, PR China
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in The Central Nervous System, Ministry of EducationTianjin City, No. 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Yi Kang
- Department of Orthopedics, Tianjin Medical University General HospitalNo. 154 Anshan Road, Heping District, Tianjin 300052, PR China
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in The Central Nervous System, Ministry of EducationTianjin City, No. 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Lu Liu
- Department of Orthopedics, Tianjin Medical University General HospitalNo. 154 Anshan Road, Heping District, Tianjin 300052, PR China
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in The Central Nervous System, Ministry of EducationTianjin City, No. 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Yongfu Lou
- Department of Orthopedics, Tianjin Medical University General HospitalNo. 154 Anshan Road, Heping District, Tianjin 300052, PR China
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in The Central Nervous System, Ministry of EducationTianjin City, No. 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Chi Zhang
- Department of Orthopedics, Tianjin Medical University General HospitalNo. 154 Anshan Road, Heping District, Tianjin 300052, PR China
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in The Central Nervous System, Ministry of EducationTianjin City, No. 154 Anshan Road, Heping District, Tianjin 300052, PR China
| | - Yanzheng Gao
- Department of Orthopedics, Henan Province People’s HospitalZhengzhou 450000, Henan, China
| | - Xiaohong Kong
- School of Medicine, Nankai UniversityNo. 94 Weijin Road, Nankai District, Tianjin 300071, PR China
| | - Shiqing Feng
- Department of Orthopedics, Tianjin Medical University General HospitalNo. 154 Anshan Road, Heping District, Tianjin 300052, PR China
- Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neurorepair and Regeneration in The Central Nervous System, Ministry of EducationTianjin City, No. 154 Anshan Road, Heping District, Tianjin 300052, PR China
| |
Collapse
|
35
|
Sanchez-Rojas L, Gómez-Pinedo U, Benito-Martin MS, León-Espinosa G, Rascón-Ramirez F, Lendinez C, Martínez-Ramos C, Matías-Guiu J, Pradas MM, Barcia JA. Biohybrids of scaffolding hyaluronic acid biomaterials plus adipose stem cells home local neural stem and endothelial cells: Implications for reconstruction of brain lesions after stroke. J Biomed Mater Res B Appl Biomater 2018; 107:1598-1606. [PMID: 30307108 DOI: 10.1002/jbm.b.34252] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/23/2018] [Accepted: 09/08/2018] [Indexed: 12/25/2022]
Abstract
Endogenous neurogenesis in stroke is insufficient to replace the lost brain tissue, largely due to the lack of a proper biological structure to let new cells dwell in the damaged area. We hypothesized that scaffolds made of hyaluronic acid (HA) biomaterials (BM) could provide a suitable environment to home not only new neurons, but also vessels, glia and neurofilaments. Further, the addition of exogenous cells, such as adipose stem cells (ASC) could increase this effect. Athymic mice were randomly assigned to a one of four group: stroke alone, stroke and implantation of BM, stroke and implantation of BM with ASC, and sham operated animals. Stroke model consisted of middle cerebral artery thrombosis with FeCl3 . After 30 days, animals underwent magnetic resonance imaging (MRI) and were sacrificed. Proliferation and neurogenesis increased at the subventricular zone ipsilateral to the ventricle and neuroblasts, glial, and endothelial cells forming capillaries were seen inside the BM. Those effects increased when ASC were added, while there was less inflammatory reaction. Three-dimensional scaffolds made of HA are able to home newly formed neurons, glia, and endothelial cells permitting the growth neurofilaments inside them. The addition of ASC increase these effects and decrease the inflammatory reaction to the implant. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 1598-1606, 2019.
Collapse
Affiliation(s)
- Leyre Sanchez-Rojas
- Laboratorio de Medicina Regenerativa, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | - Ulises Gómez-Pinedo
- Laboratorio de Medicina Regenerativa, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | - María Soledad Benito-Martin
- Laboratorio de Medicina Regenerativa, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | - Gonzalo León-Espinosa
- Laboratorio de Medicina Regenerativa, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain.,Instituto Cajal, CSIC; Laboratorio Cajal de Circuitos Corticales (CTB), Universidad Politécnica de Madrid; Facultad de Farmacia, Universidad San Pablo CEU, Madrid, Spain
| | - Fernando Rascón-Ramirez
- Servicio de Neurocirugía, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Cristina Lendinez
- Laboratorio de Medicina Regenerativa, Instituto de Neurociencias, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | - Cristina Martínez-Ramos
- Center for Biomaterials and Tissue Engineering, Universitat Politécnica de Valencia, C. de Vera s/n, Valencia, Spain
| | - Jorge Matías-Guiu
- Servicio de Neurología. Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| | - Manuel Monleón Pradas
- Center for Biomaterials and Tissue Engineering, Universitat Politécnica de Valencia, C. de Vera s/n, Valencia, Spain.,Networking Research Center on Bioengineering, Biomaterials and Nanomedicine, Valencia, Spain
| | - Juan A Barcia
- Servicio de Neurocirugía, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
36
|
Wu S, FitzGerald KT, Giordano J. On the Viability and Potential Value of Stem Cells for Repair and Treatment of Central Neurotrauma: Overview and Speculations. Front Neurol 2018; 9:602. [PMID: 30150968 PMCID: PMC6099099 DOI: 10.3389/fneur.2018.00602] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 07/06/2018] [Indexed: 12/12/2022] Open
Abstract
Central neurotrauma, such as spinal cord injury or traumatic brain injury, can damage critical axonal pathways and neurons and lead to partial to complete loss of neural function that is difficult to address in the mature central nervous system. Improvement and innovation in the development, manufacture, and delivery of stem-cell based therapies, as well as the continued exploration of newer forms of stem cells, have allowed the professional and public spheres to resolve technical and ethical questions that previously hindered stem cell research for central nervous system injury. Recent in vitro and in vivo models have demonstrated the potential that reprogrammed autologous stem cells, in particular, have to restore functionality and induce regeneration-while potentially mitigating technical issues of immunogenicity, rejection, and ethical issues of embryonic derivation. These newer stem-cell based approaches are not, however, without concerns and problems of safety, efficacy, use and distribution. This review is an assessment of the current state of the science, the potential solutions that have been and are currently being explored, and the problems and questions that arise from what appears to be a promising way forward (i.e., autologous stem cell-based therapies)-for the purpose of advancing the research for much-needed therapeutic interventions for central neurotrauma.
Collapse
Affiliation(s)
- Samantha Wu
- Pellegrino Center for Clinical Bioethics, Georgetown University Medical Center, Washington, DC, United States
| | - Kevin T. FitzGerald
- Pellegrino Center for Clinical Bioethics, Georgetown University Medical Center, Washington, DC, United States
- Department of Oncology, Georgetown University Medical Center, Washington, DC, United States
| | - James Giordano
- Pellegrino Center for Clinical Bioethics, Georgetown University Medical Center, Washington, DC, United States
- Departments of Neurology and Biochemistry, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
37
|
Radial Extracorporeal Shock Wave Therapy Enhances the Proliferation and Differentiation of Neural Stem Cells by Notch, PI3K/AKT, and Wnt/β-catenin Signaling. Sci Rep 2017; 7:15321. [PMID: 29127399 PMCID: PMC5681501 DOI: 10.1038/s41598-017-15662-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 10/20/2017] [Indexed: 01/05/2023] Open
Abstract
Neural stem cell (NSC) proliferation and differentiation play a pivotal role in the repair of brain function in central nervous system (CNS) diseases. Radial extracorporeal shock wave therapy (rESWT) is a non-invasive and innovative treatment for many conditions, yet little is known about the effects of this treatment on NSCs. Mouse NSCs (NE-4C) were exposed to rESWT with 1.0, 1.5, 2.0, 2.5, 3.0, and 3.5 bar (500 impulses, and 2 Hz) in vitro. Cell viability test results indicated that rESWT, at a dose of 2.5 bar, 500 impulses, and 2 Hz, increased NE-4C viability within 72 h, and that the PI3K/AKT pathway was involved in its mechanisms. Exposure to rESWT also affected proliferation and differentiation of NE-4C after 8 weeks, which may be associated with Wnt/β-catenin and Notch pathways. This assessment is corroborated by the ability of inhibitors of Wnt/β-catenin [Dickkopf-1 (Dkk-1)] and the Notch pathway (DAPT) to weaken proliferation and differentiation of NSCs. In summary, a proper dose of rESWT enhanced NSCs augment via the PI3K/AKT pathway initially. Also, Wnt/β-catenin and the Notch pathway play important roles in regulation of the long-term efficacy of rESWT. This study reveals a novel approach to culture NSCs in vitro and support neurogenesis.
Collapse
|