1
|
Le NTN, Han CL, Delila L, Nebie O, Chien HT, Wu YW, Buée L, Blum D, Burnouf T. Proteomics of human platelet lysates and insight from animal studies on platelet protein diffusion to hippocampus upon intranasal administration. APL Bioeng 2024; 8:026111. [PMID: 38726021 PMCID: PMC11080963 DOI: 10.1063/5.0196553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 04/23/2024] [Indexed: 05/12/2024] Open
Abstract
Human platelet lysates (HPLs) from allogeneic platelet concentrates (PCs) are biomaterials, which are rich in various trophic factors, increasingly used in regenerative medicine and biotherapy. Understanding how preparation methods influence the HPL protein profile, biological function, and clinical outcomes is crucial. Our study sheds light on the proteomes and functionality of different HPLs, with the aim of advancing their scientifically grounded clinical applications. To achieve this, PCs suspended in plasma underwent three distinct processing methods, resulting in seven HPL types. We used three characterization techniques: label-free proteomics and tandem mass tag (TMT)-based quantitative proteomics, both before and after the immunodepletion of abundant plasma proteins. Bioinformatic tools assessed the proteome, and western blotting validated our quantitative proteomics data. Subsequent pre-clinical studies with fluorescent labeling and label-free proteomics were used as a proof of concept for brain diffusion. Our findings revealed 1441 proteins detected using the label-free method, 952 proteins from the TMT experiment before and after depletion, and 1114 proteins from the subsequent TMT experiment on depleted HPLs. Most detected proteins were cytoplasmic, playing key roles in catalysis, hemostasis, and immune responses. Notably, the processing methodologies significantly influenced HPL compositions, their canonical pathways, and, consequently, their functionality. Each HPL exhibited specific abundant proteins, providing valuable insight for tailored clinical applications. Immunoblotting results for selected proteins corroborated our quantitative proteomics data. The diffusion and differential effects to the hippocampus of a neuroprotective HPL administered intranasally to mice were demonstrated. This proteomics study advances our understanding of HPLs, suggesting ways to standardize and customize their production for better clinical efficacy in regenerative medicine and biotherapy. Proteomic analyses also offered objective evidence that HPPL, upon intranasal delivery, not only effectively diffuses to the hippocampus but also alters protein expression in mice, bolstering its potential as a treatment for memory impairments.
Collapse
Affiliation(s)
- Nhi Thao Ngoc Le
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei, Taiwan
| | - Chia-Li Han
- Master Program in Clinical Genomics and Proteomics, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Liling Delila
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei, Taiwan
| | | | - Hsin-Tung Chien
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei, Taiwan
| | - Yu-Wen Wu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei, Taiwan
| | | | - David Blum
- Authors to whom correspondence should be addressed: and . Tel.: +886 988 925 235
| | - Thierry Burnouf
- Authors to whom correspondence should be addressed: and . Tel.: +886 988 925 235
| |
Collapse
|
2
|
Alito A, Fontana JM, Franzini Tibaldeo E, Verme F, Piterà P, Miller E, Cremascoli R, Brioschi A, Capodaglio P. Whole-Body Cryostimulation in Multiple Sclerosis: A Scoping Review. J Clin Med 2024; 13:2003. [PMID: 38610768 PMCID: PMC11012586 DOI: 10.3390/jcm13072003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/15/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Multiple sclerosis (MS) is the most common cause of non-traumatic long-term disability in young adults. Whole-body cryostimulation (WBC) is a cold-based physical therapy known to induce physiological exercise-mimicking changes in the cardiovascular, neuromuscular, immune, and endocrine systems and to influence functional and psychological parameters by exposing the human body to cryogenic temperatures (≤-110 °C) for 2-3 min. The purpose of this scoping review is to present an overall view on the potential role of WBC as an adjuvant therapy in the treatment of MS. PubMed, ScienceDirect, Embase, and Web of Science were searched up to 30 November 2023, and a total of 13 articles were included. WBC may have beneficial antioxidant effects as a short-term adjuvant treatment in MS. There were no significant changes in antioxidant enzymes, nitric oxide levels, metalloproteinase levels, blood counts, rheology, and biochemistry. WBC can lead to a reduction in fatigue and an improvement in functional status, with a significant effect on both mental and physical well-being. There were no reported adverse effects. The results suggest that WBC may complement therapeutic options for patients with MS, as the effects of cryogenic cold stimulation have been shown to activate antioxidant processes and improve functional status, mood, anxiety, and fatigue.
Collapse
Affiliation(s)
- Angelo Alito
- Department of Biomedical, Dental Sciences and Morphological and Functional Images, University of Messina, 98125 Messina, Italy;
| | - Jacopo Maria Fontana
- IRCCS, Istituto Auxologico Italiano, Orthopedic Rehabilitation Unit, Research Laboratory in Biomechanics and Rehabilitation, San Giuseppe Hospital, Piancavallo, 28921 Verbania, Italy; (J.M.F.); (P.C.)
| | - Eleonora Franzini Tibaldeo
- Department of Surgical Sciences, Physical and Rehabilitation Medicine, University of Torino, 10121 Torino, Italy;
| | - Federica Verme
- IRCCS, Istituto Auxologico Italiano, Orthopedic Rehabilitation Unit, Research Laboratory in Biomechanics and Rehabilitation, San Giuseppe Hospital, Piancavallo, 28921 Verbania, Italy; (J.M.F.); (P.C.)
| | - Paolo Piterà
- Department of Clinical and Biological Sciences, University of Turin, 10043 Torino, Italy;
| | - Elzbieta Miller
- Department of Neurological Rehabilitation, Medical University of Lodz, Milionowa 14, 93-113 Lodz, Poland;
| | - Riccardo Cremascoli
- IRCCS, Istituto Auxologico Italiano, Unit of Neurology and Neurorehabilitation, San Giuseppe Hospital, Piancavallo, 28921 Verbania, Italy; (R.C.); (A.B.)
| | - Andrea Brioschi
- IRCCS, Istituto Auxologico Italiano, Unit of Neurology and Neurorehabilitation, San Giuseppe Hospital, Piancavallo, 28921 Verbania, Italy; (R.C.); (A.B.)
| | - Paolo Capodaglio
- IRCCS, Istituto Auxologico Italiano, Orthopedic Rehabilitation Unit, Research Laboratory in Biomechanics and Rehabilitation, San Giuseppe Hospital, Piancavallo, 28921 Verbania, Italy; (J.M.F.); (P.C.)
- Department of Surgical Sciences, Physical and Rehabilitation Medicine, University of Torino, 10121 Torino, Italy;
| |
Collapse
|
3
|
Paris A, Lakatos A. Cell and gene therapy for amyotrophic lateral sclerosis. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:217-241. [PMID: 39341656 DOI: 10.1016/b978-0-323-90120-8.00017-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal and incurable neurodegenerative disorder with rapidly progressive skeletal muscle weakness, which can also cause a variable cognitive deficit. Genetic causes are only identified in approximately 10% of all cases, with complex genotype-phenotype associations, making it challenging to identify treatment targets. What further hampers therapeutic development is a broad heterogeneity in mechanisms, possible targets, and disturbances across various cell types, aside from the cortical and spinal motor neurons that lie at the heart of the pathology of ALS. Over the last decade, significant progress in biotechnologic techniques, cell and ribonucleic acid (RNA) engineering, animal models, and patient-specific human stem cell and organoid models have accelerated both mechanistic and therapeutic discoveries. The growing number of clinical trials mirrors this. This chapter reviews the current state of human preclinical models supporting trial strategies as well as recent clinical cell and gene therapy approaches.
Collapse
Affiliation(s)
- Alvar Paris
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom; Department of Neurology, Cambridge University Hospitals NHS Trust, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - András Lakatos
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom; Department of Neurology, Cambridge University Hospitals NHS Trust, Addenbrooke's Hospital, Cambridge, United Kingdom.
| |
Collapse
|
4
|
Albini M, Krawczun-Rygmaczewska A, Cesca F. Astrocytes and brain-derived neurotrophic factor (BDNF). Neurosci Res 2023; 197:42-51. [PMID: 36780947 DOI: 10.1016/j.neures.2023.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 01/17/2023] [Accepted: 02/02/2023] [Indexed: 02/13/2023]
Abstract
Astrocytes are emerging in the neuroscience field as crucial modulators of brain functions, from the molecular control of synaptic plasticity to orchestrating brain-wide circuit activity for cognitive processes. The cellular pathways through which astrocytes modulate neuronal activity and plasticity are quite diverse. In this review, we focus on neurotrophic pathways, mostly those mediated by brain-derived neurotrophic factor (BDNF). Neurotrophins are a well-known family of trophic factors with pleiotropic functions in neuronal survival, maturation and activity. Within the brain, BDNF is the most abundantly expressed and most studied of all neurotrophins. While we have detailed knowledge of the effect of BDNF on neurons, much less is known about its physiology on astroglia. However, over the last years new findings emerged demonstrating that astrocytes take an active part into BDNF physiology. In this work, we discuss the state-of-the-art knowledge about astrocytes and BDNF. Indeed, astrocytes sense extracellular BDNF through its specific TrkB receptors and activate intracellular responses that greatly vary depending on the brain area, stage of development and receptors expressed. Astrocytes also uptake and recycle BDNF / proBDNF at synapses contributing to synaptic plasticity. Finally, experimental evidence is now available describing deficits in astrocytic BDNF in several neuropathologies, suggesting that astrocytic BDNF may represent a promising target for clinical translation.
Collapse
Affiliation(s)
- Martina Albini
- Department of Experimental Medicine, University of Genova, Italy; IIT Center for Synaptic Neuroscience and Technology, Genova, Italy
| | - Alicja Krawczun-Rygmaczewska
- IIT Center for Synaptic Neuroscience and Technology, Genova, Italy; Department of Life Sciences, University of Trieste, Italy
| | - Fabrizia Cesca
- IIT Center for Synaptic Neuroscience and Technology, Genova, Italy; Department of Life Sciences, University of Trieste, Italy.
| |
Collapse
|
5
|
Rizzuti M, Sali L, Melzi V, Scarcella S, Costamagna G, Ottoboni L, Quetti L, Brambilla L, Papadimitriou D, Verde F, Ratti A, Ticozzi N, Comi GP, Corti S, Gagliardi D. Genomic and transcriptomic advances in amyotrophic lateral sclerosis. Ageing Res Rev 2023; 92:102126. [PMID: 37972860 DOI: 10.1016/j.arr.2023.102126] [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: 06/01/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/19/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder and the most common motor neuron disease. ALS shows substantial clinical and molecular heterogeneity. In vitro and in vivo models coupled with multiomic techniques have provided important contributions to unraveling the pathomechanisms underlying ALS. To date, despite promising results and accumulating knowledge, an effective treatment is still lacking. Here, we provide an overview of the literature on the use of genomics, epigenomics, transcriptomics and microRNAs to deeply investigate the molecular mechanisms developing and sustaining ALS. We report the most relevant genes implicated in ALS pathogenesis, discussing the use of different high-throughput sequencing techniques and the role of epigenomic modifications. Furthermore, we present transcriptomic studies discussing the most recent advances, from microarrays to bulk and single-cell RNA sequencing. Finally, we discuss the use of microRNAs as potential biomarkers and promising tools for molecular intervention. The integration of data from multiple omic approaches may provide new insights into pathogenic pathways in ALS by shedding light on diagnostic and prognostic biomarkers, helping to stratify patients into clinically relevant subgroups, revealing novel therapeutic targets and supporting the development of new effective therapies.
Collapse
Affiliation(s)
- Mafalda Rizzuti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Sali
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Melzi
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Simone Scarcella
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy
| | - Gianluca Costamagna
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy
| | - Linda Ottoboni
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy
| | - Lorenzo Quetti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenzo Brambilla
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Federico Verde
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy; Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Antonia Ratti
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy; Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Nicola Ticozzi
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy; Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Giacomo Pietro Comi
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy; Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Corti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy.
| | - Delia Gagliardi
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
6
|
Valerio LSA, Carrick FR, Bedoya L, Sreerama S, Sugaya K. Neural Differentiation of Induced Pluripotent Stem Cells for a Xenogeneic Material-Free 3D Neurological Disease Model Neurulation from Pluripotent Cells Using a Human Hydrogel. Curr Issues Mol Biol 2023; 45:4574-4588. [PMID: 37367039 DOI: 10.3390/cimb45060290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/14/2023] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
Alzheimer's Disease (AD) is characterized by synapse and neuronal loss and the accumulation of neurofibrillary tangles and Amyloid β plaques. Despite significant research efforts to understand the late stages of the disease, its etiology remains largely unknown. This is in part because of the imprecise AD models in current use. In addition, little attention has been paid to neural stem cells (NSC), which are the cells responsible for the development and maintenance of brain tissue during an individual's lifespan. Thus, an in vitro 3D human brain tissue model using induced pluripotent stem (iPS) cell-derived neural cells in human physiological conditions may be an excellent alternative to standard models to investigate AD pathology. Following the differentiation process mimicking development, iPS cells can be turned into NSCs and, ultimately, neural cells. During differentiation, the traditionally used xenogeneic products may alter the cells' physiology and prevent accurate disease pathology modeling. Hence, establishing a xenogeneic material-free cell culture and differentiation protocol is essential. This study investigated the differentiation of iPS cells to neural cells using a novel extracellular matrix derived from human platelet lysates (PL Matrix). We compared the stemness properties and differentiation efficacies of iPS cells in a PL matrix against those in a conventional 3D scaffold made of an oncogenic murine-matrix. Using well-defined conditions without xenogeneic material, we successfully expanded and differentiated iPS cells into NSCs via dual-SMAD inhibition, which regulates the BMP and TGF signaling cascades in a manner closer to human conditions. This in vitro, 3D, xenogeneic-free scaffold will enhance the quality of disease modeling for neurodegenerative disease research, and the knowledge produced could be used in developing more effective translational medicine.
Collapse
Affiliation(s)
- Luis Sebastian Alexis Valerio
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- Institute for Scientific Research and Technology Services (INDICASAT), City of Knowledge 0801, Panama
- Department of Biotechnology, Acharya Nagarjuna University, Guntur 522510, India
| | - Frederick Robert Carrick
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- MGH Institute of Health Professions, Boston, MA 02129, USA
- Centre for Mental Health Research in Association, University of Cambridge, Cambridge CB2 1TN, UK
- Department of Neurology, Carrick Institute, Cape Canaveral, FL 32920, USA
| | - Lina Bedoya
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Sandeep Sreerama
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Kiminobu Sugaya
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
- Institute for Scientific Research and Technology Services (INDICASAT), City of Knowledge 0801, Panama
- Department of Biotechnology, Acharya Nagarjuna University, Guntur 522510, India
| |
Collapse
|
7
|
Najafi S, Najafi P, Kaffash Farkhad N, Hosseini Torshizi G, Assaran Darban R, Boroumand AR, Sahab-Negah S, Khodadoust MA, Tavakol-Afshari J. Mesenchymal stem cell therapy in amyotrophic lateral sclerosis (ALS) patients: A comprehensive review of disease information and future perspectives. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:872-881. [PMID: 37427325 PMCID: PMC10329242 DOI: 10.22038/ijbms.2023.66364.14572] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 03/15/2023] [Indexed: 07/11/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a rare deadly progressive neurological disease that primarily affects the upper and lower motor neurons with an annual incidence rate of 0.6 to 3.8 per 100,000 people. Weakening and gradual atrophy of the voluntary muscles are the first signs of the disease onset affecting all aspects of patients' lives, including eating, speaking, moving, and even breathing. Only 5-10% of patients have a familial type of the disease and show an autosomal dominant pattern, but the cause of the disease is unknown in the remaining 90% of patients (Sporadic ALS). However, in both types of disease, the patient's survival is 2 to 5 years from the disease onset. Some clinical and molecular biomarkers, magnetic resonance imaging (MRI), blood or urine test, muscle biopsy, and genetic testing are complementary methods for disease diagnosis. Unfortunately, with the exception of Riluzole, the only medically approved drug for the management of this disease, there is still no definitive cure for it. In this regard, the use of mesenchymal stem cells (MSCs) for the treatment or management of the disease has been common in preclinical and clinical studies for many years. MSCs are multipotent cells having immunoregulatory, anti-inflammatory, and differentiation ability that makes them a good candidate for this purpose. This review article aims to discuss multiple aspects of ALS disease and focus on MSCs' role in disease management based on performed clinical trials.
Collapse
Affiliation(s)
- Shahrzad Najafi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Parizad Najafi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Najmeh Kaffash Farkhad
- Immunology Research Center, Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Reza Assaran Darban
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Amir Reza Boroumand
- Neuroscience Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sajad Sahab-Negah
- Neuroscience Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran
| | - Mohammad Ali Khodadoust
- Immunology Research Center, Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jalil Tavakol-Afshari
- Immunology Research Center, Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
8
|
Delila L, Nebie O, Le NTN, Barro L, Chou M, Wu Y, Watanabe N, Takahara M, Buée L, Blum D, Devos D, Burnouf T. Neuroprotective activity of a virus-safe nanofiltered human platelet lysate depleted of extracellular vesicles in Parkinson's disease and traumatic brain injury models. Bioeng Transl Med 2023; 8:e10360. [PMID: 36684076 PMCID: PMC9842020 DOI: 10.1002/btm2.10360] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/15/2022] [Accepted: 06/07/2022] [Indexed: 01/25/2023] Open
Abstract
Brain administration of human platelet lysates (HPL) is a potential emerging biotherapy of neurodegenerative and traumatic diseases of the central nervous system. HPLs being prepared from pooled platelet concentrates, thereby increasing viral risks, manufacturing processes should incorporate robust virus-reduction treatments. We evaluated a 19 ± 2-nm virus removal nanofiltration process using hydrophilic regenerated cellulose hollow fibers on the properties of a neuroprotective heat-treated HPL (HPPL). Spiking experiments demonstrated >5.30 log removal of 20-22-nm non-enveloped minute virus of mice-mock particles using an immuno-quantitative polymerase chain reaction assay. The nanofiltered HPPL (NHPPL) contained a range of neurotrophic factors like HPPL. There was >2 log removal of extracellular vesicles (EVs), associated with decreased expression of pro-thrombogenic phosphatidylserine and procoagulant activity. LC-MS/MS proteomics showed that ca. 80% of HPPL proteins, including neurotrophins, cytokines, and antioxidants, were still found in NHPPL, whereas proteins associated with some infections and cancer-associated pathways, pro-coagulation and EVs, were removed. NHPPL maintained intact neuroprotective activity in Lund human mesencephalic dopaminergic neuron model of Parkinson's disease (PD), stimulated the differentiation of SH-SY5Y neuronal cells and showed preserved anti-inflammatory function upon intranasal administration in a mouse model of traumatic brain injury (TBI). Therefore, nanofiltration of HPL is feasible, lowers the viral, prothrombotic and procoagulant risks, and preserves the neuroprotective and anti-inflammatory properties in neuronal pre-clinical models of PD and TBI.
Collapse
Affiliation(s)
- Liling Delila
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical EngineeringTaipei Medical UniversityTaipeiTaiwan
| | - Ouada Nebie
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical EngineeringTaipei Medical UniversityTaipeiTaiwan
- Univ. Lille, Inserm, CHU‐Lille, U1172, Lille Neuroscience & CognitionLilleFrance
- Alzheimer & TauopathiesLabex DISTALZLilleFrance
| | - Nhi Thao Ngoc Le
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical EngineeringTaipei Medical UniversityTaipeiTaiwan
| | - Lassina Barro
- International PhD Program in Biomedical Engineering, College of Biomedical EngineeringTaipei Medical UniversityTaipeiTaiwan
- Present address:
National Center of Blood TransfusionOuagadougouBurkina Faso
| | - Ming‐Li Chou
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical EngineeringTaipei Medical UniversityTaipeiTaiwan
- Present address:
Institute of Clinical Medicine, National Yang Ming Chiao Tung UniversityTaipeiTaiwan
| | - Yu‐Wen Wu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical EngineeringTaipei Medical UniversityTaipeiTaiwan
| | | | | | - Luc Buée
- Univ. Lille, Inserm, CHU‐Lille, U1172, Lille Neuroscience & CognitionLilleFrance
- Alzheimer & TauopathiesLabex DISTALZLilleFrance
- NeuroTMULilleLille Neuroscience & CognitionLilleFrance
| | - David Blum
- Univ. Lille, Inserm, CHU‐Lille, U1172, Lille Neuroscience & CognitionLilleFrance
- Alzheimer & TauopathiesLabex DISTALZLilleFrance
- NeuroTMULilleLille Neuroscience & CognitionLilleFrance
| | - David Devos
- Univ. Lille, Inserm, CHU‐Lille, U1172, Lille Neuroscience & CognitionLilleFrance
- NeuroTMULilleLille Neuroscience & CognitionLilleFrance
| | - Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical EngineeringTaipei Medical UniversityTaipeiTaiwan
- International PhD Program in Biomedical Engineering, College of Biomedical EngineeringTaipei Medical UniversityTaipeiTaiwan
- NeuroTMULilleTaipei Medical UniversityTaipeiTaiwan
- International PhD Program in Cell Therapy and Regeneration MedicineTaipei Medical UniversityTaipeiTaiwan
- PhD Program in Graduate Institute of Mind Brain and Consciousness, College of Humanities and Social SciencesTaipei Medical UniversityTaipeiTaiwan
- Neuroscience Research CenterTaipei Medical UniversityTaipeiTaiwan
| |
Collapse
|
9
|
The multifaceted role of platelets in mediating brain function. Blood 2022; 140:815-827. [PMID: 35609283 PMCID: PMC9412009 DOI: 10.1182/blood.2022015970] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 05/11/2022] [Indexed: 11/30/2022] Open
Abstract
Platelets, the small, anucleate blood cells that originate from megakaryocytes in the bone marrow, are typically associated with coagulation. However, it is now apparent that platelets are more multifaceted than originally thought, with their function extending beyond their traditional role in hemostasis to acting as important mediators of brain function. In this review, we outline the broad repertoire of platelet function in the central nervous system, focusing on the similarities between platelets and neurons. We also summarize the role that platelets play in the pathophysiology of various neurological diseases, with a particular focus on neuroinflammation and neurodegeneration. Finally, we highlight the exciting prospect of harnessing the unique features of the platelet proteome and extracellular vesicles, which are rich in neurotrophic, antioxidative, and antiinflammatory factors, for the development of novel neuroprotective and neuroregenerative interventions to treat various neurodegenerative and traumatic pathologies.
Collapse
|
10
|
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating motor neuron disease for which there is currently no robust therapy. Recent progress in understanding ALS disease mechanisms and genetics in combination with innovations in gene modulation strategies creates promising new options for the development of ALS therapies. In recent years, six gene modulation therapies have been tested in ALS patients. These target gain-of-function pathology of the most common ALS genes, SOD1, C9ORF72, FUS, and ATXN2, using adeno-associated virus (AAV)-mediated microRNAs and antisense oligonucleotides (ASOs). Here, we review the latest clinical and preclinical advances in gene modulation approaches for ALS, including gene silencing, gene correction, and gene augmentation. These techniques have the potential to positively impact the direction of future research trials and transform ALS treatments for this grave disease.
Collapse
Affiliation(s)
- Katharina E Meijboom
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Robert H Brown
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
11
|
Nebie O, Buée L, Blum D, Burnouf T. Can the administration of platelet lysates to the brain help treat neurological disorders? Cell Mol Life Sci 2022; 79:379. [PMID: 35750991 PMCID: PMC9243829 DOI: 10.1007/s00018-022-04397-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/09/2022] [Accepted: 05/23/2022] [Indexed: 11/03/2022]
Abstract
Neurodegenerative disorders of the central nervous system (CNS) and brain traumatic insults are characterized by complex overlapping pathophysiological alterations encompassing neuroinflammation, alterations of synaptic functions, oxidative stress, and progressive neurodegeneration that eventually lead to irreversible motor and cognitive dysfunctions. A single pharmacological approach is unlikely to provide a complementary set of molecular therapeutic actions suitable to resolve these complex pathologies. Recent preclinical data are providing evidence-based scientific rationales to support biotherapies based on administering neurotrophic factors and extracellular vesicles present in the lysates of human platelets collected from healthy donors to the brain. Here, we present the most recent findings on the composition of the platelet proteome that can activate complementary signaling pathways in vivo to trigger neuroprotection, synapse protection, anti-inflammation, antioxidation, and neurorestoration. We also report experimental data where the administration of human platelet lysates (HPL) was safe and resulted in beneficial neuroprotective effects in established rodent models of neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, traumatic brain injury, and stroke. Platelet-based biotherapies, prepared from collected platelet concentrates (PC), are emerging as a novel pragmatic and accessible translational therapeutic strategy for treating neurological diseases. Based on this assumption, we further elaborated on various clinical, manufacturing, and regulatory issues that need to be addressed to ensure the ethical supply, quality, and safety of HPL preparations for treating neurodegenerative and traumatic pathologies of the CNS. HPL made from PC may become a unique approach for scientifically based treatments of neurological disorders readily accessible in low-, middle-, and high-income countries.
Collapse
Affiliation(s)
- Ouada Nebie
- College of Biomedical Engineering, Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience and Cognition, 59045, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, LiCEND, 59000, Lille, France
| | - Luc Buée
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience and Cognition, 59045, Lille, France
- Alzheimer and Tauopathies, LabEx DISTALZ, LiCEND, 59000, Lille, France
- NeuroTMULille International Laboratory, Univ. Lille, Lille, France
| | - David Blum
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience and Cognition, 59045, Lille, France.
- Alzheimer and Tauopathies, LabEx DISTALZ, LiCEND, 59000, Lille, France.
- NeuroTMULille International Laboratory, Univ. Lille, Lille, France.
- NeuroTMULille International Laboratory, Taipei Medical University, Taipei, 11031, Taiwan.
| | - Thierry Burnouf
- College of Biomedical Engineering, Graduate Institute of Biomedical Materials and Tissue Engineering, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan.
- NeuroTMULille International Laboratory, Taipei Medical University, Taipei, 11031, Taiwan.
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan.
- International PhD Program in Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.
- Brain and Consciousness Research Centre, Taipei Medical University Shuang-Ho Hospital, New Taipei City, 23561, Taiwan.
- Neuroscience Research Center, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
12
|
Bangari DS, Lanigan LG, Goulet F, Siso S, Bolon B. Society of Toxicologic Pathology Neuropathology Interest Group Article: Neuropathologic Findings in Nonhuman Primates Associated With Administration of Biomolecule-Based Test Articles. Toxicol Pathol 2022; 50:693-711. [PMID: 35695393 DOI: 10.1177/01926233221101314] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The increasing specificity of novel druggable targets coupled with the complexity of emerging therapeutic modalities for treating human diseases has created a growing need for nonhuman primates (NHPs) as models for translational drug discovery and nonclinical safety assessment. In particular, NHPs are critical for investigating potential unexpected/undesired on-target and off-target liabilities associated with administration of candidate biotherapeutics (nucleic acids, proteins, viral gene therapy vectors, etc.) to treat nervous system disorders. Nervous system findings unique to or overrepresented in NHPs administered biomolecule-based ("biologic") test articles include mononuclear cell infiltration in most neural tissues for all biomolecule classes as well as neuronal necrosis with glial cell proliferation in sensory ganglia for certain viral vectors. Such test article-related findings in NHPs often must be differentiated from procedural effects (e.g., local parenchymal or meningeal reactions associated with an injection site or implanted catheter to administer a test article directly into the central nervous system) or spontaneous background findings (e.g., neuronal autophagy in sensory ganglia).
Collapse
Affiliation(s)
- Dinesh S Bangari
- Global Discovery Pathology, Translational In-Vivo Models Platform, Sanofi, Cambridge, Massachusetts, USA
| | | | - Felix Goulet
- Charles River Laboratories, Senneville, Quebec, Canada
| | - Silvia Siso
- Translational Imaging and Pathology, Codiak BioSciences, Cambridge, Massachusetts, USA
| | | |
Collapse
|
13
|
Banerjee P, Elliott E, Rifai OM, O'Shaughnessy J, McDade K, Abrahams S, Chandran S, Smith C, Gregory JM. NLRP3 inflammasome as a key molecular target underlying cognitive resilience in amyotrophic lateral sclerosis. J Pathol 2022; 256:262-268. [PMID: 34883532 DOI: 10.1002/path.5846] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/05/2021] [Accepted: 12/06/2021] [Indexed: 11/09/2022]
Abstract
Up to 50% of amyotrophic lateral sclerosis patients present with cognitive deficits in addition to motor dysfunction, but the molecular mechanisms underlying diverse clinical and pathological presentations remain poorly understood. There is therefore an unmet need to identify molecular drivers of cognitive dysfunction to enable better therapeutic targeting and prognostication. To address this, we employed a non-biased approach to identify molecular targets using a deeply phenotyped, clinically stratified cohort of cognitively affected and unaffected brain regions from three brain regions of 13 amyotrophic lateral sclerosis patients with the same cognitive screening test performed during life. Using NanoString molecular barcoding as a sensitive mRNA sequencing technique on post-mortem tissue, we profiled a data-driven panel of 770 genes using the Neuropathology Panel, followed by region and cell type-specific validation using BaseScope in situ hybridisation and immunohistochemistry. We identified 50 significantly dysregulated genes that are distinct between cognitively affected and unaffected brain regions. Using BaseScope in situ hybridisation, we also demonstrate that macromolecular complex regulation, notably NLRP3 inflammasome modulation, is a potential, therapeutically targetable, pathological correlate of cognitive resilience in ALS. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Poulomi Banerjee
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
- The Euan MacDonald Centre, University of Edinburgh, Edinburgh, UK
| | - Elizabeth Elliott
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
- The Euan MacDonald Centre, University of Edinburgh, Edinburgh, UK
| | - Olivia M Rifai
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
- The Euan MacDonald Centre, University of Edinburgh, Edinburgh, UK
| | - Judi O'Shaughnessy
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Karina McDade
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Sharon Abrahams
- The Euan MacDonald Centre, University of Edinburgh, Edinburgh, UK
- School of Philosophy, Psychology and Language Science, University of Edinburgh, Edinburgh, UK
- The Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, UK
| | - Siddharthan Chandran
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
- The Euan MacDonald Centre, University of Edinburgh, Edinburgh, UK
- The Anne Rowling Regenerative Neurology Clinic, University of Edinburgh, Edinburgh, UK
| | - Colin Smith
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- The Euan MacDonald Centre, University of Edinburgh, Edinburgh, UK
| | - Jenna M Gregory
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
- UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
- The Euan MacDonald Centre, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
14
|
Yan L, Guo MS, Zhang Y, Yu L, Wu JM, Tang Y, Ai W, Zhu FD, Law BYK, Chen Q, Yu CL, Wong VKW, Li H, Li M, Zhou XG, Qin DL, Wu AG. Dietary Plant Polyphenols as the Potential Drugs in Neurodegenerative Diseases: Current Evidence, Advances, and Opportunities. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5288698. [PMID: 35237381 PMCID: PMC8885204 DOI: 10.1155/2022/5288698] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 01/10/2022] [Accepted: 01/28/2022] [Indexed: 02/07/2023]
Abstract
Neurodegenerative diseases, including Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD), are characterized by the progressive degeneration of neurons. Although the etiology and pathogenesis of neurodegenerative diseases have been studied intensively, the mechanism is still in its infancy. In general, most neurodegenerative diseases share common molecular mechanisms, and multiple risks interact and promote the pathologic process of neurogenerative diseases. At present, most of the approved drugs only alleviate the clinical symptoms but fail to cure neurodegenerative diseases. Numerous studies indicate that dietary plant polyphenols are safe and exhibit potent neuroprotective effects in various neurodegenerative diseases. However, low bioavailability is the biggest obstacle for polyphenol that largely limits its adoption from evidence into clinical practice. In this review, we summarized the widely recognized mechanisms associated with neurodegenerative diseases, such as misfolded proteins, mitochondrial dysfunction, oxidative damage, and neuroinflammatory responses. In addition, we summarized the research advances about the neuroprotective effect of the most widely reported dietary plant polyphenols. Moreover, we discussed the current clinical study and application of polyphenols and the factors that result in low bioavailability, such as poor stability and low permeability across the blood-brain barrier (BBB). In the future, the improvement of absorption and stability, modification of structure and formulation, and the combination therapy will provide more opportunities from the laboratory into the clinic for polyphenols. Lastly, we hope that the present review will encourage further researches on natural dietary polyphenols in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Lu Yan
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Min-Song Guo
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Yue Zhang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Lu Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Jian-Ming Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Yong Tang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Wei Ai
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Feng-Dan Zhu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Betty Yuen-Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Qi Chen
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
- Department of Nursing, Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chong-Lin Yu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Vincent Kam-Wai Wong
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - Hua Li
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Mao Li
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Xiao-Gang Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| | - Da-Lian Qin
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macau SAR, China
| | - An-Guo Wu
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability Evaluation, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy; Education Ministry Key Laboratory of Medical Electrophysiology, College of Preclinical Medicine, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
15
|
Burnouf T, Goubran HA. Regenerative effect of expired platelet concentrate in human therapy: An update. Transfus Apher Sci 2022; 61:103363. [DOI: 10.1016/j.transci.2022.103363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
16
|
Whole and fractionated human platelet lysate biomaterials-based biotherapy induces strong neuroprotection in experimental models of amyotrophic lateral sclerosis. Biomaterials 2021; 280:121311. [PMID: 34952382 DOI: 10.1016/j.biomaterials.2021.121311] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 10/26/2021] [Accepted: 12/03/2021] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease of motor neurons leading to death within 3 years and without a curative treatment. Neurotrophic growth factors (NTFs) are pivotal for cell survival. A reason for the lack of patient efficacy with single recombinant NTF brain infusion is likely to be due to the synergistic neuroprotective action of multiple NTFs on a diverse set of signaling pathways. Fractionated (protein size <50, <30, <10, <3 kDa) heat-treated human platelet lysate (HHPL) preparations were adapted for use in brain tissue with the aim of demonstrating therapeutic value in ALS models and further elucidation of the mechanisms of action. In neuronal culture all fractions induced Akt-dependent neuroprotection as well as a strong anti-apoptotic and anti-ferroptotic action. In the <3 kDa fraction anti-ferroptotic properties were shown to be GPX4 dependent highlighting a role for other platelet elements associated with NTFs. In the SOD1G86R mouse model, lifespan was strongly increased by intracerebroventricular delivery of HHPL or by intranasal administration of <3 kDa fraction. Our results suggest that the platelet lysate biomaterials are neuroprotective in ALS. Further studies would now validate theragnostic biomarker on its antiferroptotic action, for further clinical development.
Collapse
|
17
|
Nyam-Erdene A, Nebie O, Delila L, Buée L, Devos D, Chou SY, Blum D, Burnouf T. Characterization and Chromatographic Isolation of Platelet Extracellular Vesicles from Human Platelet Lysates for Applications in Neuroregenerative Medicine. ACS Biomater Sci Eng 2021; 7:5823-5835. [PMID: 34846835 DOI: 10.1021/acsbiomaterials.1c01226] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Human platelet lysates (HPLs) made from clinical-grade platelet concentrates are currently evaluated in the preclinical models of Parkinson's disease, Alzheimer's disease, traumatic brain injury, and others, as a new polyvalent neuroprotective biotherapy of the central nervous system. However, the presence and content of extracellular vesicles (EVs) in HPLs and their potential contribution to the neuroprotective and neurorestorative activities of HPLs are still unknown. We, therefore, characterized the EVs present in four different HPL preparations and after purification by size-exclusion chromatography. We then tested the effect of the isolated EVs on neuronal cell repair. We identified that all four HPLs contained a high and similar amount of EVs (1011 to 1012/mL) with a mean size ranging from ca. 50 to 300 nm and a negative zeta potential as determined by nanoparticle tracking analysis and dynamic light scattering. Western blot analysis revealed that the EVs present in HPLs expressed the clusters of differentiation 41 (CD41) and 61 (CD61) characteristic of platelets. These EVs were efficiently isolated from HPL proteins by Sepharose CL-2B size-exclusion column chromatography as confirmed by total protein determination and protein profile by sodium dodecyl sulfate polyacrylamide gel electrophoresis, with 73-85% recovery and maintenance of their size, negative zeta potential, and CD41 and CD61 expression. Interestingly, the EVs purified from the four HPLs exhibited a differential capacity to promote cell growth and migration in a wound-healing assay using SH-SY5Y neuronal cells, and one EV preparation stimulated network formation in primary neuronal cultures. These data indicated that the EVs present in HPLs have different neuroregenerative capacities and that some EV preparations may have interesting applications as a stand-alone therapy for usage in neuroregenerative medicine.
Collapse
Affiliation(s)
- Ariunjargal Nyam-Erdene
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 101, Taiwan
| | - Ouada Nebie
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Liling Delila
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan
| | - Luc Buée
- Université de Lille, Inserm U1172, CHU-Lille, Lille Neuroscience & Cognition, Lille 59000, France.,Alzheimer & Tauopathies, Labex DISTALZ, Lille 59000, France.,NeuroTMULille International Laboratory, Université de Lille, Lille 59000, France
| | - David Devos
- Université de Lille, Inserm U1172, CHU-Lille, Lille Neuroscience & Cognition, Lille 59000, France.,NeuroTMULille International Laboratory, Université de Lille, Lille 59000, France
| | - Szu-Yi Chou
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan.,NeuroTMULille International Laboratory, Taipei Medical University, Taipei 101, Taiwan
| | - David Blum
- Université de Lille, Inserm U1172, CHU-Lille, Lille Neuroscience & Cognition, Lille 59000, France.,Alzheimer & Tauopathies, Labex DISTALZ, Lille 59000, France.,NeuroTMULille International Laboratory, Université de Lille, Lille 59000, France
| | - Thierry Burnouf
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 101, Taiwan.,Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan.,NeuroTMULille International Laboratory, Taipei Medical University, Taipei 101, Taiwan.,International PhD Program in Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan.,Brain and Consciousness Research Centre, TMU Shuang Ho Hospital, New Taipei City 106, Taiwan
| |
Collapse
|
18
|
Nebie O, Carvalho K, Barro L, Delila L, Faivre E, Renn TY, Chou ML, Wu YW, Nyam-Erdene A, Chou SY, Buée L, Hu CJ, Peng CW, Devos D, Blum D, Burnouf T. Human platelet lysate biotherapy for traumatic brain injury: preclinical assessment. Brain 2021; 144:3142-3158. [PMID: 34086871 PMCID: PMC8634089 DOI: 10.1093/brain/awab205] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/13/2021] [Accepted: 05/11/2021] [Indexed: 11/24/2022] Open
Abstract
Traumatic brain injury (TBI) leads to major brain anatomopathological damages underlined by neuroinflammation, oxidative stress and progressive neurodegeneration, ultimately leading to motor and cognitive deterioration. The multiple pathological events resulting from TBI can be addressed not by a single therapeutic approach, but rather by a synergistic biotherapy capable of activating a complementary set of signalling pathways and providing synergistic neuroprotective, anti-inflammatory, antioxidative, and neurorestorative activities. Human platelet lysate might fulfil these requirements as it is composed of a plethora of biomolecules readily accessible as a TBI biotherapy. In the present study, we tested the therapeutic potential of human platelet lysate using in vitro and in vivo models of TBI. We first prepared and characterized platelet lysate from clinical-grade human platelet concentrates. Platelets were pelletized, lysed by three freeze-thaw cycles, and centrifuged. The supernatant was purified by 56°C 30 min heat treatment and spun to obtain the heat-treated platelet pellet lysate that was characterized by ELISA and proteomic analyses. Two mouse models were used to investigate platelet lysate neuroprotective potential. The injury was induced by an in-house manual controlled scratching of the animals' cortex or by controlled cortical impact injury. The platelet lysate treatment was performed by topical application of 60 µl in the lesioned area, followed by daily 60 µl intranasal administration from Day 1 to 6 post-injury. Platelet lysate proteomics identified over 1000 proteins including growth factors, neurotrophins, and antioxidants. ELISA detected several neurotrophic and angiogenic factors at ∼1-50 ng/ml levels. We demonstrate, using two mouse models of TBI, that topical application and intranasal platelet lysate consistently improved mouse motor function in the beam and rotarod tests, mitigated cortical neuroinflammation, and oxidative stress in the injury area, as revealed by downregulation of pro-inflammatory genes and the reduction in reactive oxygen species levels. Moreover, platelet lysate treatment reduced the loss of cortical synaptic proteins. Unbiased proteomic analyses revealed that heat-treated platelet pellet lysate reversed several pathways promoted by both controlled cortical impact and cortical brain scratch and related to transport, postsynaptic density, mitochondria or lipid metabolism. The present data strongly support, for the first time, that human platelet lysate is a reliable and effective therapeutic source of neurorestorative factors. Therefore, brain administration of platelet lysate is a therapeutical strategy that deserves serious and urgent consideration for universal brain trauma treatment.
Collapse
Affiliation(s)
- Ouada Nebie
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of
Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog—Lille Neuroscience and
Cognition, Lille F-59000, France
- Alzheimer and Tauopathies, LabEx DISTALZ, LiCEND, Lille F-59000,
France
| | - Kevin Carvalho
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog—Lille Neuroscience and
Cognition, Lille F-59000, France
- Alzheimer and Tauopathies, LabEx DISTALZ, LiCEND, Lille F-59000,
France
| | - Lassina Barro
- International PhD Program in Biomedical Engineering, Taipei Medical
University, Taipei, 11031, Taiwan
| | - Liling Delila
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of
Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
| | - Emilie Faivre
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog—Lille Neuroscience and
Cognition, Lille F-59000, France
- Alzheimer and Tauopathies, LabEx DISTALZ, LiCEND, Lille F-59000,
France
| | - Ting-Yi Renn
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical
University, Taipei, 11031, Taiwan
| | - Ming-Li Chou
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of
Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University,
Taipei, Taiwan
| | - Yu-Wen Wu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of
Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
| | - Ariunjargal Nyam-Erdene
- International PhD Program in Biomedical Engineering, Taipei Medical
University, Taipei, 11031, Taiwan
| | - Szu-Yi Chou
- NeuroTMULille International Laboratory, Taipei Medical
University, Taipei, 11031, Taiwan
- PhD Program for Neural Regenerative Medicine, College of Medical Science and
Technology, Taipei Medical University and National Health Research
Institutes, Taipei, 11031, Taiwan
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science
and Technology, Taipei Medical University, Taipei, 11031, Taiwan
| | - Luc Buée
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog—Lille Neuroscience and
Cognition, Lille F-59000, France
- Alzheimer and Tauopathies, LabEx DISTALZ, LiCEND, Lille F-59000,
France
- NeuroTMULille International Laboratory, Univ. Lille, Lille,
F-59000 France
| | - Chaur-Jong Hu
- NeuroTMULille International Laboratory, Taipei Medical
University, Taipei, 11031, Taiwan
- PhD Program for Neural Regenerative Medicine, College of Medical Science and
Technology, Taipei Medical University and National Health Research
Institutes, Taipei, 11031, Taiwan
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science
and Technology, Taipei Medical University, Taipei, 11031, Taiwan
- Dementia Center, Department of Neurology, Shuang Ho Hospital, Taipei Medical
University, New Taipei City, 23561, Taiwan
- Neurology, School of Medicine, College of Medicine, Taipei Medical
University, Taipei, 11031, Taiwan
| | - Chih-Wei Peng
- International PhD Program in Biomedical Engineering, Taipei Medical
University, Taipei, 11031, Taiwan
- School of Biomedical Engineering, College of Biomedical Engineering, Taipei
Medical University, Taipei, 11031, Taiwan
| | - David Devos
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog—Lille Neuroscience and
Cognition, Lille F-59000, France
- NeuroTMULille International Laboratory, Univ. Lille, Lille,
F-59000 France
| | - David Blum
- University of Lille, Inserm, CHU Lille, U1172 - LilNCog—Lille Neuroscience and
Cognition, Lille F-59000, France
- Alzheimer and Tauopathies, LabEx DISTALZ, LiCEND, Lille F-59000,
France
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science
and Technology, Taipei Medical University, Taipei, 11031, Taiwan
| | - Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of
Biomedical Engineering, Taipei Medical University, Taipei, 11031, Taiwan
- International PhD Program in Biomedical Engineering, Taipei Medical
University, Taipei, 11031, Taiwan
- Institute of Clinical Medicine, National Yang-Ming University,
Taipei, Taiwan
- School of Biomedical Engineering, College of Biomedical Engineering, Taipei
Medical University, Taipei, 11031, Taiwan
- International PhD Program in Cell Therapy and Regeneration, College of
Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- Brain and Consciousness Research Centre, Taipei Medical University Shuang Ho
Hospital, New Taipei City, 23561, Taiwan
| |
Collapse
|
19
|
Therapeutic strategies for C9orf72 amyotrophic lateral sclerosis and frontotemporal dementia. Curr Opin Neurol 2021; 34:748-755. [PMID: 34392299 PMCID: PMC8678157 DOI: 10.1097/wco.0000000000000984] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW An intronic G4C2 expansion mutation in C9orf72 is the most common genetic cause of amyotrophic lateral sclerosis and frontotemporal dementia (C9-ALS/FTD). Although there are currently no treatments for this insidious, fatal disease, intense research has led to promising therapeutic strategies, which will be discussed here. RECENT FINDINGS Therapeutic strategies for C9-ALS/FTD have primarily focused on reducing the toxic effects of mutant expansion RNAs or the dipeptide repeat proteins (DPRs). The pathogenic effects of G4C2 expansion transcripts have been targeted using approaches aimed at promoting their degradation, inhibiting nuclear export or silencing transcription. Other promising strategies include immunotherapy to reduce the DPRs themselves, reducing RAN translation, removing the repeats using DNA or RNA editing and manipulation of downstream disease-altered stress granule pathways. Finally, understanding the molecular triggers that lead to pheno-conversion may lead to opportunities that can delay symptomatic disease onset. SUMMARY A large body of evidence implicates RAN-translated DPRs as a main driver of C9-ALS/FTD. Promising therapeutic strategies for these devastating diseases are being rapidly developed with several approaches already in or approaching clinical trials.
Collapse
|
20
|
Pikatza-Menoio O, Elicegui A, Bengoetxea X, Naldaiz-Gastesi N, López de Munain A, Gerenu G, Gil-Bea FJ, Alonso-Martín S. The Skeletal Muscle Emerges as a New Disease Target in Amyotrophic Lateral Sclerosis. J Pers Med 2021; 11:671. [PMID: 34357138 PMCID: PMC8307751 DOI: 10.3390/jpm11070671] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 01/02/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder that leads to progressive degeneration of motor neurons (MNs) and severe muscle atrophy without effective treatment. Most research on ALS has been focused on the study of MNs and supporting cells of the central nervous system. Strikingly, the recent observations of pathological changes in muscle occurring before disease onset and independent from MN degeneration have bolstered the interest for the study of muscle tissue as a potential target for delivery of therapies for ALS. Skeletal muscle has just been described as a tissue with an important secretory function that is toxic to MNs in the context of ALS. Moreover, a fine-tuning balance between biosynthetic and atrophic pathways is necessary to induce myogenesis for muscle tissue repair. Compromising this response due to primary metabolic abnormalities in the muscle could trigger defective muscle regeneration and neuromuscular junction restoration, with deleterious consequences for MNs and thereby hastening the development of ALS. However, it remains puzzling how backward signaling from the muscle could impinge on MN death. This review provides a comprehensive analysis on the current state-of-the-art of the role of the skeletal muscle in ALS, highlighting its contribution to the neurodegeneration in ALS through backward-signaling processes as a newly uncovered mechanism for a peripheral etiopathogenesis of the disease.
Collapse
Affiliation(s)
- Oihane Pikatza-Menoio
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| | - Amaia Elicegui
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| | - Xabier Bengoetxea
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
| | - Neia Naldaiz-Gastesi
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| | - Adolfo López de Munain
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
- Department of Neurology, Donostialdea Integrated Health Organization, Osakidetza Basque Health Service, 20014 Donostia/San Sebastián, Spain
- Department of Neurosciences, Faculty of Medicine and Nursery, University of the Basque Country UPV-EHU, 20014 Donostia/San Sebastián, Spain
| | - Gorka Gerenu
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
- Department of Physiology, University of the Basque Country UPV-EHU, 48940 Leioa, Spain
| | - Francisco Javier Gil-Bea
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| | - Sonia Alonso-Martín
- Neuromuscular Diseases Group, Neurosciences Area, Biodonostia Health Research Institute, 20014 Donostia/San Sebastián, Spain; (O.P.-M.); (A.E.); (X.B.); (N.N.-G.); (A.L.d.M.); (G.G.); (F.J.G.-B.)
- CIBERNED, Carlos III Institute, Spanish Ministry of Economy & Competitiveness, 28031 Madrid, Spain
| |
Collapse
|
21
|
Lara-Rodarte R, Cortés D, Soriano K, Carmona F, Rocha L, Estudillo E, López-Ornelas A, Velasco I. Mouse Embryonic Stem Cells Expressing GDNF Show Enhanced Dopaminergic Differentiation and Promote Behavioral Recovery After Grafting in Parkinsonian Rats. Front Cell Dev Biol 2021; 9:661656. [PMID: 34239871 PMCID: PMC8258349 DOI: 10.3389/fcell.2021.661656] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 05/17/2021] [Indexed: 11/17/2022] Open
Abstract
Parkinson's disease (PD) is characterized by the progressive loss of midbrain dopaminergic neurons (DaNs) of the substantia nigra pars compacta and the decrease of dopamine in the brain. Grafting DaN differentiated from embryonic stem cells (ESCs) has been proposed as an alternative therapy for current pharmacological treatments. Intrastriatal grafting of such DaNs differentiated from mouse or human ESCs improves motor performance, restores DA release, and suppresses dopamine receptor super-sensitivity. However, a low percentage of grafted neurons survive in the brain. Glial cell line-derived neurotrophic factor (GDNF) is a strong survival factor for DaNs. GDNF has proved to be neurotrophic for DaNs in vitro and in vivo, and induces axonal sprouting and maturation. Here, we engineered mouse ESCs to constitutively produce human GDNF, to analyze DaN differentiation and the possible neuroprotection by transgenic GDNF after toxic challenges in vitro, or after grafting differentiated DaNs into the striatum of Parkinsonian rats. GDNF overexpression throughout in vitro differentiation of mouse ESCs increases the proportion of midbrain DaNs. These transgenic cells were less sensitive than control cells to 6-hydroxydopamine in vitro. After grafting control or GDNF transgenic DaNs in hemi-Parkinsonian rats, we observed significant recoveries in both pharmacological and non-pharmacological behavioral tests, as well as increased striatal DA release, indicating that DaNs are functional in the brain. The graft volume, the number of surviving neurons, the number of DaNs present in the striatum, and the proportion of DaNs in the grafts were significantly higher in rats transplanted with GDNF-expressing cells, when compared to control cells. Interestingly, no morphological alterations in the brain of rats were found after grafting of GDNF-expressing cells. This approach is novel, because previous works have use co-grafting of DaNs with other cell types that express GDNF, or viral transduction in the host tissue before or after grafting of DaNs. In conclusion, GDNF production by mouse ESCs contributes to enhanced midbrain differentiation and permits a higher number of surviving DaNs after a 6-hydroxydopamine challenge in vitro, as well as post-grafting in the lesioned striatum. These GDNF-expressing ESCs can be useful to improve neuronal survival after transplantation.
Collapse
Affiliation(s)
- Rolando Lara-Rodarte
- Instituto de Fisiología Celular – Neurociencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez,”Mexico City, Mexico
| | - Daniel Cortés
- Instituto de Fisiología Celular – Neurociencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez,”Mexico City, Mexico
| | - Karla Soriano
- Instituto de Fisiología Celular – Neurociencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez,”Mexico City, Mexico
| | - Francia Carmona
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados (Cinvestav), Mexico City, Mexico
| | - Luisa Rocha
- Departamento de Farmacobiología, Centro de Investigación y de Estudios Avanzados (Cinvestav), Mexico City, Mexico
| | - Enrique Estudillo
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez,”Mexico City, Mexico
| | - Adolfo López-Ornelas
- Instituto de Fisiología Celular – Neurociencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez,”Mexico City, Mexico
- División de Investigación, Hospital Juárez de México, Mexico City, Mexico
| | - Iván Velasco
- Instituto de Fisiología Celular – Neurociencias, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Laboratorio de Reprogramación Celular, Instituto Nacional de Neurología y Neurocirugía “Manuel Velasco Suárez,”Mexico City, Mexico
| |
Collapse
|
22
|
Ciervo Y, Gatto N, Allen C, Grierson A, Ferraiuolo L, Mead RJ, Shaw PJ. Adipose-derived stem cells protect motor neurons and reduce glial activation in both in vitro and in vivo models of ALS. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 21:413-433. [PMID: 33869658 PMCID: PMC8044387 DOI: 10.1016/j.omtm.2021.03.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 03/23/2021] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative condition for which new therapeutic options are urgently needed. We injected GFP+ adipose-derived stem cells (EGFP-ADSCs) directly into the cerebrospinal fluid (CSF) of transgenic SOD1G93A mice, a well-characterized model of familial ALS. Despite short-term survival of the injected cells and limited engraftment efficiency, EGFP-ADSCs improved motor function and delayed disease onset by promoting motor neuron (MN) survival and reducing glial activation. We then tested the in vitro neuroprotective potential of mouse ADSCs in astrocyte/MN co-cultures where ALS astrocytes show neurotoxicity. ADSCs were able to rescue MN death caused by ALS astrocytes derived from symptomatic SOD1G93A mice. Further, ADSCs were found to reduce the inflammatory signature of ALS astrocytes by inhibiting the release of pro-inflammatory mediators and inducing the secretion of neuroprotective factors. Finally, mouse ADSCs were able to protect MNs from the neurotoxicity mediated by human induced astrocytes (iAstrocytes) derived from patients with either sporadic or familial ALS, thus for the first time showing the potential therapeutic translation of ADSCs across the spectrum of human ALS. These data in two translational models of ALS show that, through paracrine mechanisms, ADSCs support MN survival and modulate the toxic microenvironment that contributes to neurodegeneration in ALS.
Collapse
Affiliation(s)
- Yuri Ciervo
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Faculty of Medicine, Dentistry and Health, The University of Sheffield, 385 Glossop Rd., Sheffield S10 2HQ, UK
| | - Noemi Gatto
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Faculty of Medicine, Dentistry and Health, The University of Sheffield, 385 Glossop Rd., Sheffield S10 2HQ, UK
| | - Chloe Allen
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Faculty of Medicine, Dentistry and Health, The University of Sheffield, 385 Glossop Rd., Sheffield S10 2HQ, UK
| | - Andrew Grierson
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Faculty of Medicine, Dentistry and Health, The University of Sheffield, 385 Glossop Rd., Sheffield S10 2HQ, UK
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Faculty of Medicine, Dentistry and Health, The University of Sheffield, 385 Glossop Rd., Sheffield S10 2HQ, UK
| | - Richard J. Mead
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Faculty of Medicine, Dentistry and Health, The University of Sheffield, 385 Glossop Rd., Sheffield S10 2HQ, UK
- Corresponding author: Richard J. Mead, PhD, Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Faculty of Medicine, Dentistry and Health, The University of Sheffield, 385 Glossop Rd., Sheffield S10 2HQ, UK.
| | - Pamela J. Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Faculty of Medicine, Dentistry and Health, The University of Sheffield, 385 Glossop Rd., Sheffield S10 2HQ, UK
- Corresponding author: Pamela J. Shaw, Professor, Dame, Sheffield Institute for Translational Neuroscience (SITraN), Department of Neuroscience, Faculty of Medicine, Dentistry and Health, The University of Sheffield, 385 Glossop Rd., Sheffield S10 2HQ, UK.
| |
Collapse
|
23
|
Puranik N, Yadav D, Chauhan PS, Kwak M, Jin JO. Exploring the Role of Gene Therapy for Neurological Disorders. Curr Gene Ther 2021; 21:11-22. [PMID: 32940177 DOI: 10.2174/1566523220999200917114101] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/30/2020] [Accepted: 08/10/2020] [Indexed: 11/22/2022]
Abstract
Gene therapy is one of the frontier fields of medical breakthroughs that poses as an effective solution to previously incurable diseases. The delivery of the corrective genetic material or a therapeutic gene into the cell restores the missing gene function and cures a plethora of diseases, incurable by the conventional medical approaches. This discovery holds the potential to treat many neurodegenerative disorders such as muscular atrophy, multiple sclerosis, Parkinson's disease (PD) and Alzheimer's disease (AD), among others. Gene therapy proves as a humane, cost-effective alternative to the exhaustive often arduous and timely impossible process of finding matched donors and extensive surgery. It also overcomes the shortcoming of conventional methods to cross the blood-brain barrier. However, the use of gene therapy is only possible after procuring the in-depth knowledge of the immuno-pathogenesis and molecular mechanism of the disease. The process of gene therapy can be broadly categorized into three main steps: elucidating the target gene, culling the appropriate vector, and determining the best mode of transfer; each step mandating pervasive research. This review aims to dissertate and summarize the role, various vectors and methods of delivery employed in gene therapy with special emphasis on therapy directed at the central nervous system (CNS) associated with neurodegenerative diseases.
Collapse
Affiliation(s)
- Nidhi Puranik
- Biological Science Department, Bharathiar University, Coimbatore, Tamil Nadu-641046, India
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea
| | - Pallavi Singh Chauhan
- Amity Institute of Biotechnology, Amity University, Gwalior, Madhya Pradesh 474005, India
| | - Minseok Kwak
- Department of Chemistry, Pukyong National University, Busan, South Korea
| | - Jun-O Jin
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan 38541, South Korea
| |
Collapse
|
24
|
Neurotrophins Time Point Intervention after Traumatic Brain Injury: From Zebrafish to Human. Int J Mol Sci 2021; 22:ijms22041585. [PMID: 33557335 PMCID: PMC7915547 DOI: 10.3390/ijms22041585] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 01/25/2021] [Accepted: 02/02/2021] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) remains the leading cause of long-term disability, which annually involves millions of individuals. Several studies on mammals reported that neurotrophins could play a significant role in both protection and recovery of function following neurodegenerative diseases such as stroke and TBI. This protective role of neurotrophins after an event of TBI has also been reported in the zebrafish model. Nevertheless, reparative mechanisms in mammalian brain are limited, and newly formed neurons do not survive for a long time. In contrast, the brain of adult fish has high regenerative properties after brain injury. The evident differences in regenerative properties between mammalian and fish brain have been ascribed to remarkable different adult neurogenesis processes. However, it is not clear if the specific role and time point contribution of each neurotrophin and receptor after TBI is conserved during vertebrate evolution. Therefore, in this review, I reported the specific role and time point of intervention for each neurotrophic factor and receptor after an event of TBI in zebrafish and mammals.
Collapse
|
25
|
Cappella M, Pradat PF, Querin G, Biferi MG. Beyond the Traditional Clinical Trials for Amyotrophic Lateral Sclerosis and The Future Impact of Gene Therapy. J Neuromuscul Dis 2021; 8:25-38. [PMID: 33074186 PMCID: PMC7902976 DOI: 10.3233/jnd-200531] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating and incurable motor neuron (MN) disorder affecting both upper and lower MNs. Despite impressive advances in the understanding of the disease’s pathological mechanism, classical pharmacological clinical trials failed to provide an efficient cure for ALS over the past twenty years. Two different gene therapy approaches were recently approved for the monogenic disease Spinal muscular atrophy, characterized by degeneration of lower MNs. This milestone suggests that gene therapy-based therapeutic solutions could be effective for the treatment of ALS. This review summarizes the possible reasons for the failure of traditional clinical trials for ALS. It provides then a focus on the advent of gene therapy approaches for hereditary forms of ALS. Specifically, it describes clinical use of antisense oligonucleotides in three familial forms of ALS, caused by mutations in SOD1, C9orf72 and FUS genes, respectively.. Clinical and pre-clinical studies based on AAV-mediated gene therapy approaches for both familial and sporadic ALS cases are presented as well. Overall, this overview highlights the potential of gene therapy as a transforming technology that will have a huge impact on treatment perspective for ALS patients and on the design of future clinical trials.
Collapse
Affiliation(s)
- Marisa Cappella
- INSERM, Institute of Myology, Centre of Research in Myology, Sorbonne Université, Paris, France
| | - Pierre-François Pradat
- INSERM, CNRS, Laboratoire d'Imagerie Biomédicale, Sorbonne Université, Paris, France.,APHP, Département de Neurologie, Hôpital Pitié-Salpêtrière, Centre référent SLA, Paris, France.,Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute Ulster University, C-TRIC, Altnagelvin Hospital, Derry/Londonderry, United Kingdom
| | - Giorgia Querin
- INSERM, Institute of Myology, Centre of Research in Myology, Sorbonne Université, Paris, France.,Association Institut de Myologie, Plateforme Essais Cliniques Adultes, Paris, France.,APHP, Service de Neuromyologie, Hôpital Pitié-Salpêtrière, Paris, France
| | - Maria Grazia Biferi
- INSERM, Institute of Myology, Centre of Research in Myology, Sorbonne Université, Paris, France
| |
Collapse
|
26
|
Abstract
Platelets play a crucial role in hemostasis, tissue regeneration and host defense. Based on these settings, platelet-rich plasma (PRP) and its derivatives are therapeutically used to promote wound healing in several scenarios. This review summarizes the biological mechanisms underlying the most traditional as well as innovative applications of PRP in wound healing. These mechanisms involve the combined action of platelet-derived growth factors and cytokines, together with the role of plasma-derived fibrillar, antioxidant and homeostatic factors. In addition, regenerative treatments with PRP consist of personalized and non-standardized methods. Thus, the quality of PRP varies depending on endogenous factors (e.g., age; gender; concomitant medication; disease-associated systemic factors; nutrition) and exogenous factors (anticoagulants and cellular composition). This review also analyses whether these factors affect the biological mechanisms of PRP in wound healing applications.
Collapse
Affiliation(s)
- Paula Oneto
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET, National Academy of Medicine, Buenos Aires, Argentina
| | - Julia Etulain
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET, National Academy of Medicine, Buenos Aires, Argentina
| |
Collapse
|
27
|
Delila L, Wu YW, Nebie O, Widyaningrum R, Chou ML, Devos D, Burnouf T. Extensive characterization of the composition and functional activities of five preparations of human platelet lysates for dedicated clinical uses. Platelets 2020; 32:259-272. [PMID: 33245683 DOI: 10.1080/09537104.2020.1849603] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Human platelet lysates (HPLs), rich in various growth factors and cell growth-promoting molecules, encompass a new range of blood products that are being used for regenerative medicine, cell therapies, and tissue engineering. Well-characterized dedicated preparations, tailor-made to best fit specific therapeutic applications, are needed for optimal clinical efficacy and safety. Here, five types of HPL were prepared from the same platelet concentrates and extensively characterized to determine and compare their proteins, growth factors, cytokines, biochemical profiles, thrombin-generating capacities, thrombin-associated proteolytic activities, phospholipid-associated procoagulant potential, contents of extracellular vesicles expressing phosphatidylserine and tissue factor, and antioxidative properties. Our results revealed that all five HPL preparations contained detectable supraphysiological levels, in the ca. 0.1 ~ 350-ng/ml range, of all growth factors assessed, except insulin-like growth factor-1 detected only in HPL containing plasma. There were significant differences observed among these HPLs in total protein content, fibrinogen, complement components C3 and C4, albumin, and immunoglobulin G, and, most importantly, in their functional coagulant and procoagulant activities and antioxidative capacities. Our data revealed that the biochemical and functional properties of HPL preparations greatly vary depending upon their mode of production, with potential impacts on the safety and efficacy for certain clinical indications. Modes of preparation of HPLs should be carefully designed, and the product properties carefully evaluated based on the intended therapeutic use to ensure optimal clinical outcomes.
Collapse
Affiliation(s)
- Liling Delila
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Yu-Wen Wu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Ouada Nebie
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Rifa Widyaningrum
- International PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ming-Li Chou
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - David Devos
- Univ. Lille, CHU-Lille, Inserm, U1172, Lille Neuroscience & Cognition, France
| | - Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.,International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.,Research Center of Biomedical Devices, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.,International PhD Program in Cell Therapy and Regeneration Medicine, College of Medicine Taipei Medical University, Taipei, Taiwan.,PhD Program in Graduate Institute of Mind Brain and Consciousness, College of Humanities and Social Sciences, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
28
|
Leyton-Jaimes MF, Ivert P, Hoeber J, Han Y, Feiler A, Zhou C, Pankratova S, Shoshan-Barmatz V, Israelson A, Kozlova EN. Empty mesoporous silica particles significantly delay disease progression and extend survival in a mouse model of ALS. Sci Rep 2020; 10:20675. [PMID: 33244084 PMCID: PMC7691331 DOI: 10.1038/s41598-020-77578-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 11/11/2020] [Indexed: 12/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating incurable neurological disorder characterized by motor neuron (MN) death and muscle dysfunction leading to mean survival time after diagnosis of only 2-5 years. A potential ALS treatment is to delay the loss of MNs and disease progression by the delivery of trophic factors. Previously, we demonstrated that implanted mesoporous silica nanoparticles (MSPs) loaded with trophic factor peptide mimetics support survival and induce differentiation of co-implanted embryonic stem cell (ESC)-derived MNs. Here, we investigate whether MSP loaded with peptide mimetics of ciliary neurotrophic factor (Cintrofin), glial-derived neurotrophic factor (Gliafin), and vascular endothelial growth factor (Vefin1) injected into the cervical spinal cord of mutant SOD1 mice affect disease progression and extend survival. We also transplanted boundary cap neural crest stem cells (bNCSCs) which have been shown previously to have a positive effect on MN survival in vitro and in vivo. We show that mimetic-loaded MSPs and bNCSCs significantly delay disease progression and increase survival of mutant SOD1 mice, and also that empty particles significantly improve the condition of ALS mice. Our results suggest that intraspinal delivery of MSPs is a potential therapeutic approach for the treatment of ALS.
Collapse
Affiliation(s)
- Marcel F Leyton-Jaimes
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel
| | - Patrik Ivert
- Department of Neuroscience, Regenerative Neurobiology, Uppsala University Biomedical Center, Box 593, 751 24, Uppsala, Sweden
| | - Jan Hoeber
- Department of Neuroscience, Regenerative Neurobiology, Uppsala University Biomedical Center, Box 593, 751 24, Uppsala, Sweden.,Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Uppsala University, Box 815, 751 08, Uppsala, Sweden
| | - Yilin Han
- Department of Neuroscience, Regenerative Neurobiology, Uppsala University Biomedical Center, Box 593, 751 24, Uppsala, Sweden
| | - Adam Feiler
- Nanologica AB, Forskargatan 20G, 151 36, Södertälje, Sweden.,Chemistry Department, KTH, Royal Institute of Technology, 100 44, Stockholm, Sweden
| | - Chunfang Zhou
- Chemistry Department, KTH, Royal Institute of Technology, 100 44, Stockholm, Sweden
| | - Stanislava Pankratova
- Laboratory of Neural Plasticity, Department of Neuroscience, University of Copenhagen, 2200, Copenhagen, Denmark.,Comparative Pediatrics and Nutrition, Department of Veterinary and Animal Sciences, University of Copenhagen, 1870, Frederiksberg C, Denmark
| | - Varda Shoshan-Barmatz
- Department of Life Sciences, The National Institute for Biotechnology in the Negev Ltd, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Adrian Israelson
- Department of Physiology and Cell Biology, Zlotowski Center for Neuroscience, Faculty of Health Sciences, Ben-Gurion University of the Negev, P.O.B. 653, 84105, Beer Sheva, Israel.
| | - Elena N Kozlova
- Department of Neuroscience, Regenerative Neurobiology, Uppsala University Biomedical Center, Box 593, 751 24, Uppsala, Sweden.
| |
Collapse
|
29
|
Molecular Mechanism of Platelet-Derived Growth Factor (PDGF)-BB-Mediated Protection Against MPP+ Toxicity in SH-SY5Y Cells. J Mol Neurosci 2020; 71:1131-1143. [DOI: 10.1007/s12031-020-01735-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 10/02/2020] [Indexed: 12/21/2022]
|
30
|
Wang J, Hu W, Feng Z, Feng M. BDNF-overexpressing human umbilical cord mesenchymal stem cell-derived motor neurons improve motor function and prolong survival in amyotrophic lateral sclerosis mice. Neurol Res 2020; 43:199-209. [PMID: 33076784 DOI: 10.1080/01616412.2020.1834775] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVE To investigate the beneficial effect of brain-derived neurotrophic factor (BDNF) -overexpressing human umbilical cord mesenchymal stem cell (hUC-MSC)-derived motor neurons in the human Cu, Zn-superoxide dismutase1 (hSOD1)G93A amyotrophic lateral sclerosis (ALS) mice. METHODS The BDNF gene was transfected into hUC-MSC-derived motor neurons by the lentivirus-mediated method. hSOD1G93A mice were assigned to the ALS, ALS/MN, and ALS/MN-BDNF groups, and intrathecally administrated phosphate-buffered saline (PBS), motor neurons, or motor neurons overexpressing BDNF, respectively. The control group included non-transgenic wild-type littermates administrated PBS. One month after transplantation, the motor function of the mice was assessed by the rotarod test, and the lumbar enlargements were then isolated to detect the expression of hSOD1 and BDNF by western blotting, and the expression of choline acetyltransferase (ChAT), homeobox protein 9 (HB9), major histocompatibility complex I (MHCI) and microtubule-associated protein-2 (MAP-2) by immunofluorescence assay. RESULTS After transplantation, mice in the ALS/MN-BDNF and ALS/MN groups both exhibited longer latency to fall and longer survival than those in the ALS group (P < 0.01 vs. P < 0.05), and the improvement was more significant in the former than in the latter. However, cell transplantation did not delay disease onset. In the lumbar enlargements of the ALS/MN-BDNF and ALS/MN groups, the expression of hSOD1 was slightly reduced without statistical significance (P > 0.05), but the expression of BDNF, ChAT and HB9, and the co-expression of MHCI and MAP-2 were significantly greater than in the ALS group (P < 0.01), with the differences also being more prominent in the former group than in the latter. CONCLUSIONS Transplantation of BDNF-overexpressing hUC-MSC-derived motor neurons can improve motor performance and prolong the survival of hSOD1G93A mice. Combining stem cell-derived motor neurons with BDNF might provide a new therapeutic strategy for ALS.
Collapse
Affiliation(s)
- Jie Wang
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University , Nanjing, China.,Department of Neurology, The Affiliated Jiangning Hospital of Nanjing Medical University , Nanjing, China
| | - Weiwei Hu
- Department of Geriatrics, Jinling Hospital, Medical School of Nanjing University , Nanjing, China
| | - Zehua Feng
- School of Stomatology, Nanjing Medical University , Nanjing, China
| | - Meijiang Feng
- Department of Geriatrics, The Second Affiliated Hospital, Nanjing Medical University , Nanjing, China.,Key Laboratory for Aging & Disease, Nanjing Medical University , Nanjing, China
| |
Collapse
|
31
|
Wen W, Wang Y, Li H, Xu H, Xu M, Frank JA, Ma M, Luo J. Mesencephalic Astrocyte-Derived Neurotrophic Factor (MANF) Regulates Neurite Outgrowth Through the Activation of Akt/mTOR and Erk/mTOR Signaling Pathways. Front Mol Neurosci 2020; 13:560020. [PMID: 33071755 PMCID: PMC7541815 DOI: 10.3389/fnmol.2020.560020] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/04/2020] [Indexed: 12/13/2022] Open
Abstract
Neurite outgrowth is essential for brain development and the recovery of brain injury and neurodegenerative diseases. In this study, we examined the role of the neurotrophic factor MANF in regulating neurite outgrowth. We generated MANF knockout (KO) neuro2a (N2a) cell lines using clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 and demonstrated that MANF KO N2a cells failed to grow neurites in response to RA stimulation. Using MANF siRNA, this finding was confirmed in human SH-SY5Y neuronal cell line. Nevertheless, MANF overexpression by adenovirus transduction or addition of MANF into culture media facilitated the growth of longer neurites in RA-treated N2a cells. MANF deficiency resulted in inhibition of Akt, Erk, mTOR, and P70S6, and impaired protein synthesis. MANF overexpression on the other hand facilitated the growth of longer neurites by activating Akt, Erk, mTOR, and P70S6. Pharmacological blockade of Akt, Erk or mTOR eliminated the promoting effect of MANF on neurite outgrowth. These findings suggest that MANF positively regulated neurite outgrowth by activating Akt/mTOR and Erk/mTOR signaling pathways.
Collapse
Affiliation(s)
- Wen Wen
- Department of Pathology, University of Iowa, Iowa City, IA, United States
| | - Yongchao Wang
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Hui Li
- Department of Pathology, University of Iowa, Iowa City, IA, United States
| | - Hong Xu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Mei Xu
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Jacqueline A Frank
- Department of Neurology, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Murong Ma
- Department of Pharmacology and Nutritional Sciences, University of Kentucky College of Medicine, Lexington, KY, United States
| | - Jia Luo
- Department of Pathology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
32
|
Volonté C, Morello G, Spampinato AG, Amadio S, Apolloni S, D’Agata V, Cavallaro S. Omics-based exploration and functional validation of neurotrophic factors and histamine as therapeutic targets in ALS. Ageing Res Rev 2020; 62:101121. [PMID: 32653439 DOI: 10.1016/j.arr.2020.101121] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 05/27/2020] [Accepted: 07/07/2020] [Indexed: 12/13/2022]
Abstract
A plethora of genetic and molecular mechanisms have been implicated in the pathophysiology of the heterogeneous and multifactorial amyotrophic lateral sclerosis (ALS) disease, and hence the conventional "one target-one drug" paradigm has failed so far to provide effective therapeutic solutions, precisely because of the complex nature of ALS. This review intends to highlight how the integration of emerging "omics" approaches may provide a rational foundation for the comprehensive exploration of molecular pathways and dynamic interactions involved in ALS, for the identification of candidate targets and biomarkers that will assist in the rapid diagnosis and prognosis, lastly for the stratification of patients into different subgroups with the aim of personalized therapeutic strategies. To this purpose, particular emphasis will be placed on some potential therapeutic targets, including neurotrophic factors and histamine signaling that both have emerged as dysregulated at different omics levels in specific subgroups of ALS patients, and have already shown promising results in in vitro and in vivo models of ALS. To conclude, we will discuss about the utility of using integrated omics coupled with network-based approaches to provide additional guidance for personalization of medicine applications in ALS.
Collapse
|
33
|
Raffa P, Scattolini V, Gerli MFM, Perin S, Cui M, De Coppi P, Elvassore N, Caccin P, Luni C, Urciuolo A. Decellularized skeletal muscles display neurotrophic effects in three-dimensional organotypic cultures. Stem Cells Transl Med 2020; 9:1233-1243. [PMID: 32578968 PMCID: PMC7519766 DOI: 10.1002/sctm.20-0090] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 05/01/2020] [Accepted: 05/07/2020] [Indexed: 12/13/2022] Open
Abstract
Skeletal muscle decellularization allows the generation of natural scaffolds that retain the extracellular matrix (ECM) mechanical integrity, biological activity, and three‐dimensional (3D) architecture of the native tissue. Recent reports showed that in vivo implantation of decellularized muscles supports muscle regeneration in volumetric muscle loss models, including nervous system and neuromuscular junctional homing. Since the nervous system plays pivotal roles during skeletal muscle regeneration and in tissue homeostasis, support of reinnervation is a crucial aspect to be considered. However, the effect of decellularized muscles on reinnervation and on neuronal axon growth has been poorly investigated. Here, we characterized residual protein composition of decellularized muscles by mass spectrometry and we show that scaffolds preserve structural proteins of the ECM of both skeletal muscle and peripheral nervous system. To investigate whether decellularized scaffolds could per se attract neural axons, organotypic sections of spinal cord were cultured three dimensionally in vitro, in presence or in absence of decellularized muscles. We found that neural axons extended from the spinal cord are attracted by the decellularized muscles and penetrate inside the scaffolds upon 3D coculture. These results demonstrate that decellularized scaffolds possess intrinsic neurotrophic properties, supporting their potential use for the treatment of clinical cases where extensive functional regeneration of the muscle is required.
Collapse
Affiliation(s)
- Paolo Raffa
- Veneto Institute of Molecular Medicine, Padova, Italy.,Women's and Children's Health Department, University of Padova, Padova, Italy
| | - Valentina Scattolini
- Veneto Institute of Molecular Medicine, Padova, Italy.,Women's and Children's Health Department, University of Padova, Padova, Italy
| | | | - Silvia Perin
- University College London Great Ormond Street Institute of Child Health, London, UK
| | - Meihua Cui
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, People's Republic of China
| | - Paolo De Coppi
- University College London Great Ormond Street Institute of Child Health, London, UK
| | - Nicola Elvassore
- Veneto Institute of Molecular Medicine, Padova, Italy.,University College London Great Ormond Street Institute of Child Health, London, UK.,Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, People's Republic of China.,Industrial Engineering Department, University of Padova, Padova, Italy
| | - Paola Caccin
- Biomedical Science Department, University of Padova, Padova, Italy
| | - Camilla Luni
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai, People's Republic of China
| | - Anna Urciuolo
- University College London Great Ormond Street Institute of Child Health, London, UK.,Institute of Pediatric Research (IRP), Fondazione Città della Speranza, Padova, Italy
| |
Collapse
|
34
|
Liscic RM, Alberici A, Cairns NJ, Romano M, Buratti E. From basic research to the clinic: innovative therapies for ALS and FTD in the pipeline. Mol Neurodegener 2020; 15:31. [PMID: 32487123 PMCID: PMC7268618 DOI: 10.1186/s13024-020-00373-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 03/27/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and Frontotemporal Degeneration (FTD) are neurodegenerative disorders, related by deterioration of motor and cognitive functions and short survival. Aside from cases with an inherited pathogenic mutation, the causes of the disorders are still largely unknown and no effective treatment currently exists. It has been shown that FTD may coexist with ALS and this overlap occurs at clinical, genetic, and molecular levels. In this work, we review the main pathological aspects of these complex diseases and discuss how the integration of the novel pathogenic molecular insights and the analysis of molecular interaction networks among all the genetic players represents a critical step to shed light on discovering novel therapeutic strategies and possibly tailoring personalized medicine approaches to specific ALS and FTD patients.
Collapse
Affiliation(s)
- Rajka Maria Liscic
- Department of Neurology, Johannes Kepler University, Linz, Austria
- School of Medicine, University of Osijek, Osijek, Croatia
| | - Antonella Alberici
- Neurology Unit, Department of Neurological Sciences and Vision, ASST-Spedali Civili-University of Brescia, Brescia, Italy
| | - Nigel John Cairns
- College of Medicine and Health and Living Systems Institute, University of Exeter, Exeter, UK
| | - Maurizio Romano
- Department of Life Sciences, Via Valerio 28, University of Trieste, 34127, Trieste, Italy
| | - Emanuele Buratti
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano 99, 34149, Trieste, Italy.
| |
Collapse
|
35
|
Nebie O, Barro L, Wu YW, Knutson F, Buée L, Devos D, Peng CW, Blum D, Burnouf T. Heat-treated human platelet pellet lysate modulates microglia activation, favors wound healing and promotes neuronal differentiation in vitro. Platelets 2020; 32:226-237. [PMID: 32106742 DOI: 10.1080/09537104.2020.1732324] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The neurorestorative efficacy of human platelet lysates in neurodegenerative disorders is still under investigation. Platelets prepared from standard and pathogen reduced platelet concentrates were pelletized, washed, concentrated, and subjected to freeze-thawing. The lysate was heated to 56°C for 30 min and characterized. Toxicity was evaluated using SH-SY5Y neuroblastoma, BV-2 microglial, and EA-hy926 endothelial cells. Inflammatory activity was tested by examining tumor necrosis factor (TNF) and cyclooxygenase (COX)-2 expressions by BV-2 microglia with or without stimulation by lipopolysaccharides (LPS). The capacity to stimulate wound healing was evaluated by a scratch assay, and the capacity to differentiate SH-SY5Y into neurons was also examined. Platelet lysates contained a range of neurotrophins. They were not toxic to SH-SY5Y, EA-hy926, or BV-2 cells, did not induce the expression of TNF or COX-2 inflammatory markers by BV-2 microglia, and decreased inflammation after LPS stimulation. They stimulated the wound closure in the scratch assay and induced SH-SY5Y differentiation as revealed by the increased length of neurites as well as β3-tubulin and neurofilament staining. These data confirm the therapeutic potential of platelet lysates in the treatment of disorders of the central nervous system and support further evaluation as novel neurorestorative biotherapy in preclinical models.
Collapse
Affiliation(s)
- Ouada Nebie
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Lassina Barro
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Yu-Wen Wu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Folke Knutson
- Clinical Immunology and Transfusion Medicine IGP, Uppsala University, Uppsala, Sweden
| | - Luc Buée
- Univ. Lille, Inserm, CHU-Lille, U1172, Lille Neuroscience & Cognition, France
| | - David Devos
- Univ. Lille, Inserm, CHU-Lille, U1172, Lille Neuroscience & Cognition, France
| | - Chih-Wei Peng
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.,School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - David Blum
- Univ. Lille, Inserm, CHU-Lille, U1172, Lille Neuroscience & Cognition, France
| | - Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.,International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan.,International PhD Program in Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan.,PhD Program in Mind, Brain & Consciousness, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
36
|
ALSUntangled 53: Carnitine supplements. Amyotroph Lateral Scler Frontotemporal Degener 2020; 21:477-483. [PMID: 32046513 DOI: 10.1080/21678421.2020.1726565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
37
|
Nebie O, Devos D, Vingtdeux V, Barro L, Devedjian JC, Jonneaux A, Chou ML, Bordet R, Buée L, Knutson F, Blum D, Burnouf T. The neuroprotective activity of heat-treated human platelet lysate biomaterials manufactured from outdated pathogen-reduced (amotosalen/UVA) platelet concentrates. J Biomed Sci 2019; 26:89. [PMID: 31666073 PMCID: PMC6822406 DOI: 10.1186/s12929-019-0579-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 10/09/2019] [Indexed: 12/11/2022] Open
Abstract
Background Effective neurorestorative therapies of neurodegenerative diseases must be developed. There is increasing interest in using human platelet lysates, rich in neurotrophic factors, as novel disease-modifying strategy of neurodegeneration. To ensure virus safety, pathogen reduction treatments should be incorporated in the preparation process of the platelet concentrates used as source material. We therefore investigated whether platelet concentrates (PC) pathogen-inactivated using a licensed photo-inactivation treatment combining photosensitive psoralen (amotosalen) and UVA irradiation (Intercept) can serve as source material to prepare platelet lysates with preserved neuroprotective activity in Parkinson’s disease models. Methods Intercept treated-PCs were centrifuged, when reaching expiry day (7 days after collection), to remove plasma and platelet additive solution. The platelet pellet was re-suspended and concentrated in phosphate buffer saline, subjected to 3 freeze-thaw cycles (− 80 °C/37 °C) then centrifuged to remove cell debris. The supernatant was recovered and further purified, or not, by heat-treatment as in our previous investigations. The content in proteins and neurotrophic factors was determined and the toxicity and neuroprotective activity of the platelet lysates towards LUHMES cells or primary cortical/hippocampal neurons were assessed using ELISA, flow cytometry, cell viability and cytotoxicity assays and proteins analysis by Western blot. Results Platelet lysates contained the expected level of total proteins (ca. 7–14 mg/mL) and neurotrophic factors. Virally inactivated and heat-treated platelet lysates did not exert detectable toxic effects on neither Lund human mesencephalic dopaminergic LUHMES cell line nor primary neurons. When used at doses of 5 and 0.5%, they enhanced the expression of tyrosine hydroxylase and neuron-specific enolase in LUHMES cells and did not significantly impact synaptic protein expression in primary neurons, respectively. Furthermore, virally-inactivated platelet lysates tested were found to exert very strong neuroprotection effects on both LUHMES and primary neurons exposed to erastin, an inducer of ferroptosis cell death. Conclusion Outdated Intercept pathogen-reduced platelet concentrates can be used to prepare safe and highly neuroprotective human heat-treated platelet pellet lysates. These data open reassuring perspectives in the possibility to develop an effective biotherapy using virally-inactivated platelet lysates rich in functional neurotrophins for neuroregenerative medicine, and for further bio-industrial development. However, the data should be confirmed in animal models. Graphical abstract ![]()
Collapse
Affiliation(s)
- Ouada Nebie
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan
| | - David Devos
- Univ Lille, Inserm, CHU Lille, UMR-S1171. Lille Neuroscience & Cognition, Degenerative and vascular cognitive disorders, F-59000, Lille, France
| | - Valérie Vingtdeux
- Univ. Lille, Inserm, CHU-Lille, UMR-S1172, Lille Neuroscience & Cognition, Alzheimer & Tauopathies, F-59000, Lille, France
| | - Lassina Barro
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Jean-Christophe Devedjian
- Univ Lille, Inserm, CHU Lille, UMR-S1171. Lille Neuroscience & Cognition, Degenerative and vascular cognitive disorders, F-59000, Lille, France
| | - Aurélie Jonneaux
- Univ Lille, Inserm, CHU Lille, UMR-S1171. Lille Neuroscience & Cognition, Degenerative and vascular cognitive disorders, F-59000, Lille, France
| | - Ming-Li Chou
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan.,Present address: INSERM UMRS 938, CdR Saint-Antoine, Laboratory Immune System, Neuroinflammation and Neurodegenerative Diseases, Saint-Antoine Hospital, Paris, France
| | - Régis Bordet
- Univ Lille, Inserm, CHU Lille, UMR-S1171. Lille Neuroscience & Cognition, Degenerative and vascular cognitive disorders, F-59000, Lille, France
| | - Luc Buée
- Univ. Lille, Inserm, CHU-Lille, UMR-S1172, Lille Neuroscience & Cognition, Alzheimer & Tauopathies, F-59000, Lille, France
| | - Folke Knutson
- Clinical Immunology and Transfusion Medicine IGP, Uppsala University, Uppsala, Sweden
| | - David Blum
- Univ. Lille, Inserm, CHU-Lille, UMR-S1172, Lille Neuroscience & Cognition, Alzheimer & Tauopathies, F-59000, Lille, France.
| | - Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan. .,International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan. .,International Ph.D. Program in Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|