1
|
Hu T, Zhou T, Goit RK, Tam KC, Chan YK, Lam WC, Lo ACY. Bioactive Glial-Derived Neurotrophic Factor from a Safe Injectable Collagen-Alginate Composite Gel Rescues Retinal Photoreceptors from Retinal Degeneration in Rabbits. Mar Drugs 2024; 22:394. [PMID: 39330275 PMCID: PMC11433152 DOI: 10.3390/md22090394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/28/2024] Open
Abstract
The management of vision-threatening retinal diseases remains challenging due to the lack of an effective drug delivery system. Encapsulated cell therapy (ECT) offers a promising approach for the continuous delivery of therapeutic agents without the need for immunosuppressants. In this context, an injectable and terminable collagen-alginate composite (CAC) ECT gel, designed with a Tet-on pro-caspase-8 system, was developed as a safe intraocular drug delivery platform for the sustained release of glial-cell-line-derived neurotrophic factor (GDNF) to treat retinal degenerative diseases. This study examined the potential clinical application of the CAC ECT gel, focusing on its safety, performance, and termination through doxycycline (Dox) administration in the eyes of healthy New Zealand White rabbits, as well as its therapeutic efficacy in rabbits with sodium-iodate (SI)-induced retinal degeneration. The findings indicated that the CAC ECT gel can be safely implanted without harming the retina or lens, displaying resistance to degradation, facilitating cell attachment, and secreting bioactive GDNF. Furthermore, the GDNF levels could be modulated by the number of implants. Moreover, Dox administration was effective in terminating gel function without causing retinal damage. Notably, rabbits with retinal degeneration treated with the gels exhibited significant functional recovery in both a-wave and b-wave amplitudes and showed remarkable efficacy in reducing photoreceptor apoptosis. Given its biocompatibility, mechanical stability, controlled drug release, terminability, and therapeutic effectiveness, our CAC ECT gel presents a promising therapeutic strategy for various retinal diseases in a clinical setting, eliminating the need for immunosuppressants.
Collapse
Affiliation(s)
- Tingyu Hu
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (T.H.); (T.Z.); (K.C.T.); (Y.K.C.); (W.-C.L.)
| | - Ting Zhou
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (T.H.); (T.Z.); (K.C.T.); (Y.K.C.); (W.-C.L.)
| | - Rajesh Kumar Goit
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (T.H.); (T.Z.); (K.C.T.); (Y.K.C.); (W.-C.L.)
- Jules Stein Eye Institute, Los Angeles, CA 90095, USA
| | - Ka Cheung Tam
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (T.H.); (T.Z.); (K.C.T.); (Y.K.C.); (W.-C.L.)
| | - Yau Kei Chan
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (T.H.); (T.Z.); (K.C.T.); (Y.K.C.); (W.-C.L.)
| | - Wai-Ching Lam
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (T.H.); (T.Z.); (K.C.T.); (Y.K.C.); (W.-C.L.)
- Department of Ophthalmology and Visual Sciences, University of British Columbia, Vancouver, BC V5Z 3N9, Canada
| | - Amy Cheuk Yin Lo
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (T.H.); (T.Z.); (K.C.T.); (Y.K.C.); (W.-C.L.)
| |
Collapse
|
2
|
Capossela L, Gatto A, Ferretti S, Di Sarno L, Graglia B, Massese M, Soligo M, Chiaretti A. Multifaceted Roles of Nerve Growth Factor: A Comprehensive Review with a Special Insight into Pediatric Perspectives. BIOLOGY 2024; 13:546. [PMID: 39056738 PMCID: PMC11273967 DOI: 10.3390/biology13070546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/11/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024]
Abstract
Nerve growth factor (NGF) is a neurotrophic peptide largely revealed for its ability to regulate the growth and survival of peripheral sensory, sympathetic, and central cholinergic neurons. The pro-survival and regenerative properties of neurotrophic factors propose a therapeutic potential in a wide range of brain diseases, and NGF, in particular, has appeared as an encouraging potential treatment. In this review, a summary of clinical studies regarding NGF and its therapeutic effects published to date, with a specific interest in the pediatric context, will be attempted. NGF has been studied in neurological disorders such as hypoxic-ischemic encephalopathy, traumatic brain injury, neurobehavioral and neurodevelopmental diseases, congenital malformations, cerebral infections, and in oncological and ocular diseases. The potential of NGF to support neuronal survival, repair, and plasticity in these contexts is highlighted. Emerging therapeutic strategies for NGF delivery, including intranasal administration as well as advanced nanotechnology-based methods, are discussed. These techniques aim to enhance NGF bioavailability and target specificity, optimizing therapeutic outcomes while minimizing systemic side effects. By synthesizing current research, this review underscores the promise and challenges of NGF-based therapies in pediatric neurology, advocating for continued innovation in delivery methods to fully harness NGF's therapeutic potential.
Collapse
Affiliation(s)
- Lavinia Capossela
- Institute of Pediatrics, Fondazione Policlinico A. Gemelli IRCCS-Università Cattolica Sacro Cuore, 00168 Rome, Italy; (S.F.); (L.D.S.); (B.G.); (A.C.)
| | - Antonio Gatto
- Institute of Pediatrics, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (A.G.); (M.M.)
| | - Serena Ferretti
- Institute of Pediatrics, Fondazione Policlinico A. Gemelli IRCCS-Università Cattolica Sacro Cuore, 00168 Rome, Italy; (S.F.); (L.D.S.); (B.G.); (A.C.)
| | - Lorenzo Di Sarno
- Institute of Pediatrics, Fondazione Policlinico A. Gemelli IRCCS-Università Cattolica Sacro Cuore, 00168 Rome, Italy; (S.F.); (L.D.S.); (B.G.); (A.C.)
| | - Benedetta Graglia
- Institute of Pediatrics, Fondazione Policlinico A. Gemelli IRCCS-Università Cattolica Sacro Cuore, 00168 Rome, Italy; (S.F.); (L.D.S.); (B.G.); (A.C.)
| | - Miriam Massese
- Institute of Pediatrics, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (A.G.); (M.M.)
| | - Marzia Soligo
- Istituto di Farmacologia Traslazionale, Consiglio Nazionale delle Ricerche (CNR), 00133 Rome, Italy;
| | - Antonio Chiaretti
- Institute of Pediatrics, Fondazione Policlinico A. Gemelli IRCCS-Università Cattolica Sacro Cuore, 00168 Rome, Italy; (S.F.); (L.D.S.); (B.G.); (A.C.)
| |
Collapse
|
3
|
Fuchs S, Caserto JS, Liu Q, Wang K, Shariati K, Hartquist CM, Zhao X, Ma M. A Glucose-Responsive Cannula for Automated and Electronics-Free Insulin Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403594. [PMID: 38639424 PMCID: PMC11223976 DOI: 10.1002/adma.202403594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/12/2024] [Indexed: 04/20/2024]
Abstract
Automated delivery of insulin based on continuous glucose monitoring is revolutionizing the way insulin-dependent diabetes is treated. However, challenges remain for the widespread adoption of these systems, including the requirement of a separate glucose sensor, sophisticated electronics and algorithms, and the need for significant user input to operate these costly therapies. Herein, a user-centric glucose-responsive cannula is reported for electronics-free insulin delivery. The cannula-made from a tough, elastomer-hydrogel hybrid membrane formed through a one-pot solvent exchange method-changes permeability to release insulin rapidly upon physiologically relevant varying glucose levels, providing simple and automated insulin delivery with no additional hardware or software. Two prototypes of the cannula are evaluated in insulin-deficient diabetic mice. The first cannula-an ends-sealed, subcutaneously inserted prototype-normalizes blood glucose levels for 3 d and controls postprandial glucose levels. The second, more translational version-a cannula with the distal end sealed and the proximal end connected to a transcutaneous injection port-likewise demonstrates tight, 3-d regulation of blood glucose levels when refilled twice daily. This proof-of-concept study may aid in the development of "smart" cannulas and next-generation insulin therapies at a reduced burden-of-care toll and cost to end-users.
Collapse
Affiliation(s)
- Stephanie Fuchs
- Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Julia S. Caserto
- Robert Frederick Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca NY, 14853, USA
| | - Qingsheng Liu
- Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Kecheng Wang
- Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Kaavian Shariati
- Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Chase M. Hartquist
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Xuanhe Zhao
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Minglin Ma
- Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| |
Collapse
|
4
|
Kavand A, Noverraz F, Gerber-Lemaire S. Recent Advances in Alginate-Based Hydrogels for Cell Transplantation Applications. Pharmaceutics 2024; 16:469. [PMID: 38675129 PMCID: PMC11053880 DOI: 10.3390/pharmaceutics16040469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
With its exceptional biocompatibility, alginate emerged as a highly promising biomaterial for a large range of applications in regenerative medicine. Whether in the form of microparticles, injectable hydrogels, rigid scaffolds, or bioinks, alginate provides a versatile platform for encapsulating cells and fostering an optimal environment to enhance cell viability. This review aims to highlight recent studies utilizing alginate in diverse formulations for cell transplantation, offering insights into its efficacy in treating various diseases and injuries within the field of regenerative medicine.
Collapse
Affiliation(s)
| | | | - Sandrine Gerber-Lemaire
- Group for Functionalized Biomaterials, Institute of Chemical Sciences and Engineering, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland; (A.K.); (F.N.)
| |
Collapse
|
5
|
Capsoni S, Cattaneo A. Getting Into the Brain: The Intranasal Approach to Enhance the Delivery of Nerve Growth Factor and Its Painless Derivative in Alzheimer’s Disease and Down Syndrome. Front Neurosci 2022; 16:773347. [PMID: 35360160 PMCID: PMC8961408 DOI: 10.3389/fnins.2022.773347] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 02/10/2022] [Indexed: 01/04/2023] Open
Abstract
The neurotrophin Nerve Growth Factor (NGF) holds a great potential as a therapeutic candidate for the treatment of neurological diseases. However, its safe and effective delivery to the brain is limited by the fact that NGF needs to be selectively targeted to the brain, to avoid severe side effects such as pain and to bypass the blood brain barrier. In this perspective, we will summarize the different approaches that have been used, or are currently applied, to deliver NGF to the brain, during preclinical and clinical trials to develop NGF as a therapeutic drug for Alzheimer’s disease. We will focus on the intranasal delivery of NGF, an approach that is used to deliver proteins to the brain in a non-invasive, safe, and effective manner minimizing systemic exposure. We will also describe the main experimental facts related to the effective intranasal delivery of a mutant form of NGF [painless NGF, human nerve growth factor painless (hNGFp)] in mouse models of Alzheimer’s disease and compare it to other ways to deliver NGF to the brain. We will also report new data on the application of intranasal delivery of hNGFp in Down Syndrome mouse model. These new data extend the therapeutic potential of hNGFp for the treatment of the dementia that is progressively associated to Down Syndrome. In conclusion, we will show how this approach can be a promising strategy and a potential solution for other unmet medical needs of safely and effectively delivering this neuroprotective neurotrophin to the brain.
Collapse
Affiliation(s)
- Simona Capsoni
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
- Section of Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
- *Correspondence: Simona Capsoni,
| | - Antonino Cattaneo
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
- European Brain Research Institute–Fondazione Rita Levi-Montalcini, Rome, Italy
| |
Collapse
|
6
|
Mesa-Infante V, Afonso-Oramas D, Salas-Hernández J, Rodríguez-Núñez J, Barroso-Chinea P. Long-term exposure to GDNF induces dephosphorylation of Ret, AKT, and ERK1/2, and is ineffective at protecting midbrain dopaminergic neurons in cellular models of Parkinson's disease. Mol Cell Neurosci 2021; 118:103684. [PMID: 34826608 DOI: 10.1016/j.mcn.2021.103684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 11/12/2021] [Accepted: 11/13/2021] [Indexed: 12/01/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) promotes differentiation, proliferation, and survival in different cell types, including dopaminergic neurons. Thus, GDNF has been proposed as a promising neuroprotective therapy in Parkinson's disease. Although findings from cellular and animal models of Parkinson's disease were encouraging, results emerging from clinical trials were not as good as expected, probably due to the inappropriate administration protocols. Despite the growing information on GDNF action mechanisms, many aspects of its pharmacological effects are still unclear and data from different studies are still contradictory. Considering that GDNF action mechanisms are mediated by its receptor tyrosine kinase Ret, which activates PI3K/AKT and MAPK/ERK signaling pathways, we aimed to investigate Ret activation and its effect over both signaling pathways in midbrain cell cultures treated with GDNF at different doses (0.3, 1, and 10 ng/ml) and times (15 min, 24 h, 24 h (7 days), and 7 continuous days). The results showed that short-term or acute (15 min, 24 h, and 24 h (7 days)) GDNF treatment in rat midbrain neurons increases Tyrosine hydroxylase (TH) expression and the phosphorylation levels of Ret (Tyr 1062), AKT (Ser 473), ERK1/2 (Thr202/Tyr204), S6 (Ser 235/236), and GSK3-β (Ser 9). However, the phosphorylation level of these kinases, TH expression, and dopamine uptake, decreased below basal levels after long-term or prolonged treatment with 1 and 10 ng/ml GDNF (7 continuous days). Our data suggest that long-term GDNF treatment inactivates the receptor by an unknown mechanism, affecting its neuroprotective capacity against degeneration caused by 6-OHDA or rotenone, while short-term exposure to GDNF promoted dopaminergic cell survival. These findings highlight the need to find new and more effective long-acting therapeutic approaches for disorders in which GDNF plays a beneficial role, including Parkinson's disease. In this regard, it is necessary to propose new GDNF treatment guidelines to regulate and control its long-term expression levels and optimize the clinical use of this trophic factor in patients with Parkinson's disease.
Collapse
Affiliation(s)
- V Mesa-Infante
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | - D Afonso-Oramas
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas de Canarias (ITB), Universidad de La Laguna, Tenerife, Spain.
| | - J Salas-Hernández
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | - J Rodríguez-Núñez
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain
| | - P Barroso-Chinea
- Departamento de Ciencias Médicas Básicas, Facultad de Medicina, Universidad de La Laguna, Tenerife, Spain; Instituto de Tecnologías Biomédicas de Canarias (ITB), Universidad de La Laguna, Tenerife, Spain.
| |
Collapse
|
7
|
Arango D, Bittar A, Esmeral NP, Ocasión C, Muñoz-Camargo C, Cruz JC, Reyes LH, Bloch NI. Understanding the Potential of Genome Editing in Parkinson's Disease. Int J Mol Sci 2021; 22:9241. [PMID: 34502143 PMCID: PMC8430539 DOI: 10.3390/ijms22179241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 01/05/2023] Open
Abstract
CRISPR is a simple and cost-efficient gene-editing technique that has become increasingly popular over the last decades. Various CRISPR/Cas-based applications have been developed to introduce changes in the genome and alter gene expression in diverse systems and tissues. These novel gene-editing techniques are particularly promising for investigating and treating neurodegenerative diseases, including Parkinson's disease, for which we currently lack efficient disease-modifying treatment options. Gene therapy could thus provide treatment alternatives, revolutionizing our ability to treat this disease. Here, we review our current knowledge on the genetic basis of Parkinson's disease to highlight the main biological pathways that become disrupted in Parkinson's disease and their potential as gene therapy targets. Next, we perform a comprehensive review of novel delivery vehicles available for gene-editing applications, critical for their successful application in both innovative research and potential therapies. Finally, we review the latest developments in CRISPR-based applications and gene therapies to understand and treat Parkinson's disease. We carefully examine their advantages and shortcomings for diverse gene-editing applications in the brain, highlighting promising avenues for future research.
Collapse
Affiliation(s)
- David Arango
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| | - Amaury Bittar
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| | - Natalia P. Esmeral
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| | - Camila Ocasión
- Grupo de Diseño de Productos y Procesos, Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (C.O.); (L.H.R.)
| | - Carolina Muñoz-Camargo
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| | - Juan C. Cruz
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| | - Luis H. Reyes
- Grupo de Diseño de Productos y Procesos, Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (C.O.); (L.H.R.)
| | - Natasha I. Bloch
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| |
Collapse
|
8
|
Amalric M, Pattij T, Sotiropoulos I, Silva JM, Sousa N, Ztaou S, Chiamulera C, Wahlberg LU, Emerich DF, Paolone G. Where Dopaminergic and Cholinergic Systems Interact: A Gateway for Tuning Neurodegenerative Disorders. Front Behav Neurosci 2021; 15:661973. [PMID: 34366802 PMCID: PMC8340002 DOI: 10.3389/fnbeh.2021.661973] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 06/14/2021] [Indexed: 12/18/2022] Open
Abstract
Historically, many investigations into neurodegenerative diseases have focused on alterations in specific neuronal populations such as, for example, the loss of midbrain dopaminergic neurons in Parkinson's disease (PD) and loss of cholinergic transmission in Alzheimer's disease (AD). However, it has become increasingly clear that mammalian brain activities, from executive and motor functioning to memory and emotional responses, are strictly regulated by the integrity of multiple interdependent neuronal circuits. Among subcortical structures, the dopaminergic nigrostriatal and mesolimbic pathways as well as cholinergic innervation from basal forebrain and brainstem, play pivotal roles in orchestrating cognitive and non-cognitive symptoms in PD and AD. Understanding the functional interactions of these circuits and the consequent neurological changes that occur during degeneration provides new opportunities to understand the fundamental inter-workings of the human brain as well as develop new potential treatments for patients with dysfunctional neuronal circuits. Here, excerpted from a session of the European Behavioral Pharmacology Society meeting (Braga, Portugal, August 2019), we provide an update on our recent work in behavioral and cellular neuroscience that primarily focuses on interactions between cholinergic and dopaminergic systems in PD models, as well as stress in AD. These brief discussions include descriptions of (1) striatal cholinergic interneurons (CINs) and PD, (2) dopaminergic and cholinergic modulation of impulse control, and (3) the use of an implantable cell-based system for drug delivery directly the into brain and (4) the mechanisms through which day life stress, a risk factor for AD, damage protein and RNA homeostasis leading to AD neuronal malfunction.
Collapse
Affiliation(s)
- Marianne Amalric
- Centre National de la Recherche Scientifique (CNRS), UMR 7291, Laboratoire de Neurosciences Cognitives, Aix-Marseille University (AMU), Marseille, France
| | - Tommy Pattij
- Amsterdam Neuroscience, Department of Anatomy and Neurosciences, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Ioannis Sotiropoulos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Braga, Portugal
| | - Joana M. Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B’s – PT Government Associate Laboratory, Braga, Portugal
| | - Nuno Sousa
- ICVS/3B’s – PT Government Associate Laboratory, Braga, Portugal
| | - Samira Ztaou
- Centre National de la Recherche Scientifique (CNRS), UMR 7291, Laboratoire de Neurosciences Cognitives, Aix-Marseille University (AMU), Marseille, France
- Department of Molecular Therapeutics, New York State Psychiatric Institute, Department of Psychiatry, Columbia University, New York, NY, United States
| | - Cristiano Chiamulera
- Department of Diagnostic and Public Health, Section of Pharmacology, University of Verona, Verona, Italy
| | | | | | - Giovanna Paolone
- Department of Diagnostic and Public Health, Section of Pharmacology, University of Verona, Verona, Italy
| |
Collapse
|
9
|
Kopach O, Pavlov AM, Sindeeva OA, Sukhorukov GB, Rusakov DA. Biodegradable Microcapsules Loaded with Nerve Growth Factor Enable Neurite Guidance and Synapse Formation. Pharmaceutics 2020; 13:E25. [PMID: 33375672 PMCID: PMC7823884 DOI: 10.3390/pharmaceutics13010025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/16/2022] Open
Abstract
Neurological disorders and traumas often involve loss of specific neuronal connections, which would require intervention with high spatial precision. We have previously demonstrated the biocompatibility and therapeutic potential of the layer-by-layer (LbL)-fabricated microcapsules aimed at the localized delivery of specific channel blockers to peripheral nerves. Here, we explore the potential of LbL-microcapsules to enable site-specific, directional action of neurotrophins to stimulate neuronal morphogenesis and synaptic circuit formation. We find that nanoengineered biodegradable microcapsules loaded with nerve growth factor (NGF) can guide the morphological development of hippocampal neurons in vitro. The presence of NGF-loaded microcapsules or their clusters increases the neurite outgrowth rate while boosting neurite branching. Microcapsule clusters appear to guide the trajectory of developing individual axons leading to the formation of functional synapses. Our observations highlight the potential of NGF-loaded, biodegradable LbL-microcapsules to help guide axonal development and possibly circuit regeneration in neuropathology.
Collapse
Affiliation(s)
- Olga Kopach
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| | - Anton M. Pavlov
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK; (A.M.P.); (O.A.S.)
- Remote Controlled Theranostic Systems Laboratory, Saratov State University, 83 Astrakhanskaya Street, 410012 Saratov, Russia
| | - Olga A. Sindeeva
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK; (A.M.P.); (O.A.S.)
- Center for Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, 3 Nobel Street, 143005 Moscow, Russia
| | - Gleb B. Sukhorukov
- School of Engineering and Materials Science, Queen Mary University of London, Mile End Road, London E1 4NS, UK; (A.M.P.); (O.A.S.)
- Center for Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, 3 Nobel Street, 143005 Moscow, Russia
| | - Dmitri A. Rusakov
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London WC1N 3BG, UK
| |
Collapse
|
10
|
Paolone G. From the Gut to the Brain and Back: Therapeutic Approaches for the Treatment of Network Dysfunction in Parkinson's Disease. Front Neurol 2020; 11:557928. [PMID: 33117258 PMCID: PMC7575743 DOI: 10.3389/fneur.2020.557928] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/04/2020] [Indexed: 12/16/2022] Open
Abstract
Parkinson's disease (PD) is a complex, multisystem, progressive, degenerative disorder characterized by severe, debilitating motor dysfunction, cognitive impairments, and mood disorders. Although preclinical research has traditionally focused on the motor deficits resulting from the loss of nigrostriatal dopaminergic neurons, up to two thirds of PD patients present separate and distinct behavioral changes. Loss of basal forebrain cholinergic neurons occurs as early as the loss of dopaminergic cells and contributes to the cognitive decline in PD. In addition, attentional deficits can limit posture control and movement efficacy caused by dopaminergic cell loss. Complicating the picture further is intracellular α-synuclein accumulation beginning in the enteric nervous system and diffusing to the substantia nigra through the dorsal motor neurons of the vagus nerve. It seems that α-synuclein's role is that of mediating dopamine synthesis, storage, and release, and its function has not been completely understood. Treating a complex, multistage network disorder, such as PD, likely requires a multipronged approach. Here, we describe a few approaches that could be used alone or perhaps in combination to achieve a greater mosaic of behavioral benefit. These include (1) using encapsulated, genetically modified cells as delivery vehicles for administering neuroprotective trophic factors, such as GDNF, in a direct and sustained means to the brain; (2) immunotherapeutic interventions, such as vaccination or the use of monoclonal antibodies against aggregated, pathological α-synuclein; (3) the continuous infusion of levodopa-carbidopa through an intestinal gel pad to attenuate the loss of dopaminergic function and manage the motor and non-motor complications in PD patients; and (4) specific rehabilitation treatment programs for drug-refractory motor complications.
Collapse
Affiliation(s)
- Giovanna Paolone
- Department of Diagnostic and Public Health - Section of Pharmacology, University of Verona, Verona, Italy
| |
Collapse
|
11
|
Salado-Manzano C, Perpiña U, Straccia M, Molina-Ruiz FJ, Cozzi E, Rosser AE, Canals JM. Is the Immunological Response a Bottleneck for Cell Therapy in Neurodegenerative Diseases? Front Cell Neurosci 2020; 14:250. [PMID: 32848630 PMCID: PMC7433375 DOI: 10.3389/fncel.2020.00250] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/17/2020] [Indexed: 12/11/2022] Open
Abstract
Neurodegenerative disorders such as Parkinson's (PD) and Huntington's disease (HD) are characterized by a selective detrimental impact on neurons in a specific brain area. Currently, these diseases have no cures, although some promising trials of therapies that may be able to slow the loss of brain cells are underway. Cell therapy is distinguished by its potential to replace cells to compensate for those lost to the degenerative process and has shown a great potential to replace degenerated neurons in animal models and in clinical trials in PD and HD patients. Fetal-derived neural progenitor cells, embryonic stem cells or induced pluripotent stem cells are the main cell sources that have been tested in cell therapy approaches. Furthermore, new strategies are emerging, such as the use of adult stem cells, encapsulated cell lines releasing trophic factors or cell-free products, containing an enriched secretome, which have shown beneficial preclinical outcomes. One of the major challenges for these potential new treatments is to overcome the host immune response to the transplanted cells. Immune rejection can cause significant alterations in transplanted and endogenous tissue and requires immunosuppressive drugs that may produce adverse effects. T-, B-lymphocytes and microglia have been recognized as the main effectors in striatal graft rejection. This review aims to summarize the preclinical and clinical studies of cell therapies in PD and HD. In addition, the precautions and strategies to ensure the highest quality of cell grafts, the lowest risk during transplantation and the reduction of a possible immune rejection will be outlined. Altogether, the wide-ranging possibilities of advanced therapy medicinal products (ATMPs) could make therapeutic treatment of these incurable diseases possible in the near future.
Collapse
Affiliation(s)
- Cristina Salado-Manzano
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedicine, University of Barcelona, Barcelona, Spain
- Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
- Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Unai Perpiña
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedicine, University of Barcelona, Barcelona, Spain
- Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
- Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | | | - Francisco J. Molina-Ruiz
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedicine, University of Barcelona, Barcelona, Spain
- Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
- Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Emanuele Cozzi
- Department of Cardio-Thoracic, Vascular Sciences and Public Health, University of Padua, Padua, Italy
- Transplant Immunology Unit, Padua University Hospital, Padua, Italy
| | - Anne E. Rosser
- Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, United Kingdom
- MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff, United Kingdom
- Brain Repair Group, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Josep M. Canals
- Laboratory of Stem Cells and Regenerative Medicine, Department of Biomedicine, University of Barcelona, Barcelona, Spain
- Production and Validation Center of Advanced Therapies (Creatio), Faculty of Medicine and Health Science, University of Barcelona, Barcelona, Spain
- Institute of Neurosciences, University of Barcelona, Barcelona, Spain
- Networked Biomedical Research Centre for Neurodegenerative Disorders (CIBERNED), Barcelona, Spain
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| |
Collapse
|
12
|
Abstract
The last decade has been a frustrating time for investigators who had envisioned major advances in the treatment of Parkinson’s disease using neurotrophic factors. The first trials of glial cell line–derived neurotrophic factor for treating Parkinson’s disease were very promising. Later blinded control trials were disappointing, not reaching the predetermined outcomes for improvement in motor function. Consideration of the problems in the studies as well as the biology of the neurotrophins used can potentially lead to more effective therapies. Parkinson’s disease presents a multitude of opportunities for the cell biologist wanting to understand its pathology and to find possible new avenues for treatment.
Collapse
|
13
|
Wahlberg LU, Emerich DF, Kordower JH, Bell W, Fradet T, Paolone G. Long-term, stable, targeted biodelivery and efficacy of GDNF from encapsulated cells in the rat and Goettingen miniature pig brain. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2020; 1:19-29. [PMID: 34909639 PMCID: PMC8663965 DOI: 10.1016/j.crphar.2020.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/19/2022] Open
Abstract
Delivering glial cell line-derived neurotrophic factor (GDNF) to the brain is a potential treatment for Parkinson's Disease (PD). Here we use an implantable encapsulated cell technology that uses modified human clonal ARPE-19 cells to deliver of GDNF to the brain. In vivo studies demonstrated sustained delivery of GDNF to the rat striatum over 6 months. Anatomical benefits and behavioral efficacy were shown in 6-OHDA lesioned rats where nigral dopaminergic neurons were preserved in neuroprotection studies and dopaminergic fibers were restored in neurorecovery studies. When larger, clinical-sized devices were implanted for 3 months into the putamen of Göttingen minipigs, GDNF was widely distributed throughout the putamen and caudate producing a significant upregulation of tyrosine hydroxylase immunohistochemistry. These results are the first to provide clear evidence that implantation of encapsulated GDNF-secreting cells deliver efficacious and biologically relevant amounts of GDNF in a sustained and targeted manner that is scalable to treat the large putamen in patients with Parkinson's disease.
Collapse
Affiliation(s)
| | | | - Jeffrey H Kordower
- Department of Neurological Sciences, Rush University Medical Center, Chicago, IL, USA
| | | | | | - Giovanna Paolone
- Department of Diagnostic and Public Health, Section of Pharmacology, University of Verona P.le, LA Scuro, Verona, Italy
| |
Collapse
|
14
|
Santos-Vizcaino E, Orive G, Pedraz JL, Hernandez RM. Clinical Applications of Cell Encapsulation Technology. Methods Mol Biol 2020; 2100:473-491. [PMID: 31939144 DOI: 10.1007/978-1-0716-0215-7_32] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cell encapsulation comprises immunoisolation three-dimensional systems for housing therapeutic cells that secrete bioactive compounds de novo and in a sustained manner. This allows transplantation of multiple allo- or xenogeneic cells without the aid of immunosuppressant drugs. Recent advances in the field have provided improvements to these cell-based drug delivery systems, which have gained the attention of the scientific community and inspired many biotechnological companies to develop their own product candidates. From micro- to macroencapsulation devices, this chapter describes some of the most important approaches that are being currently tested in late-stage clinical trials and are likely to reach the market as future game changers. Most studies involve the treatment of diabetes, eye disorders, and diseases of the central nervous system. However, many other pathologies are also amenable to benefit from this technology. Latest advances to overcome major pending challenges related to biosafety and efficacy are also discussed.
Collapse
Affiliation(s)
- Edorta Santos-Vizcaino
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain.,Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - Gorka Orive
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain.,Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain.,University Institute for Regenerative Medicine and Oral Implantology-UIRMI (UPV/EHU-Fundación Eduardo Anitua), Vitoria, Spain.,BTI Biotechnology Institute, Vitoria, Spain
| | - Jose Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain.,Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain
| | - Rosa Maria Hernandez
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz, Spain. .,Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria-Gasteiz, Spain.
| |
Collapse
|
15
|
Kopach O. Monitoring maturation of neural stem cell grafts within a host microenvironment. World J Stem Cells 2019; 11:982-989. [PMID: 31768224 PMCID: PMC6851006 DOI: 10.4252/wjsc.v11.i11.982] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2019] [Revised: 09/08/2019] [Accepted: 10/01/2019] [Indexed: 02/06/2023] Open
Abstract
Neural stem cells (NSC) act as a versatile tool for neuronal cell replacement strategies to treat neurodegenerative disorders in which functional neurorestorative mechanisms are limited. While the beneficial effects of such cell-based therapy have already been documented in terms of neurodegeneration of various origins, a neurophysiological basis for improvement in the recovery of neurological function is still not completely understood. This overview briefly describes the cumulative evidence from electrophysiological studies of NSC-derived neurons, aimed at establishing the maturation of differentiated neurons within a host microenvironment, and their integration into the host circuits, with a particular focus on the neurogenesis of NSC grafts within the post-ischemic milieu. Overwhelming evidence demonstrates that the host microenvironment largely regulates the lineage of NSC grafts. This regulatory role, as yet underestimated, raises possibilities for the favoured maturation of a subset of neural phenotypes in order to gain timely remodelling of the impaired brain tissue and amplify the therapeutic effects of NSC-based therapy for recovery of neurological function.
Collapse
Affiliation(s)
- Olga Kopach
- Department of Clinical and Experimental Epilepsy, Queen Square Institute of Neurology, University College London, London WC1 N3BG, United Kingdom.
| |
Collapse
|
16
|
Kustrimovic N, Marino F, Cosentino M. Peripheral Immunity, Immunoaging and Neuroinflammation in Parkinson's Disease. Curr Med Chem 2019; 26:3719-3753. [PMID: 30306855 DOI: 10.2174/0929867325666181009161048] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 06/26/2018] [Accepted: 09/12/2018] [Indexed: 12/20/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder among elderly population, characterized by the progressive degeneration of dopaminergic neurons in the midbrain. To date, exact cause remains unknown and the mechanism of neurons death uncertain. It is typically considered as a disease of central nervous system (CNS). Nevertheless, numerous evidence has been accumulated in several past years testifying undoubtedly about the principal role of neuroinflammation in progression of PD. Neuroinflammation is mainly associated with presence of activated microglia in brain and elevated levels of cytokine levels in CNS. Nevertheless, active participation of immune system as well has been noted, such as, elevated levels of cytokine levels in blood, the presence of auto antibodies, and the infiltration of T cell in CNS. Moreover, infiltration and reactivation of those T cells could exacerbate neuroinflammation to greater neurotoxic levels. Hence, peripheral inflammation is able to prime microglia into pro-inflammatory phenotype, which can trigger stronger response in CNS further perpetuating the on-going neurodegenerative process. In the present review, the interplay between neuroinflammation and the peripheral immune response in the pathobiology of PD will be discussed. First of all, an overview of regulation of microglial activation and neuroinflammation is summarized and discussed. Afterwards, we try to collectively analyze changes that occurs in peripheral immune system of PD patients, suggesting that these peripheral immune challenges can exacerbate the process of neuroinflammation and hence the symptoms of the disease. In the end, we summarize some of proposed immunotherapies for treatment of PD.
Collapse
Affiliation(s)
- Natasa Kustrimovic
- Center of Research in Medical Pharmacology, University of Insubria, Varese, Italy
| | - Franca Marino
- Center of Research in Medical Pharmacology, University of Insubria, Varese, Italy
| | - Marco Cosentino
- Center of Research in Medical Pharmacology, University of Insubria, Varese, Italy
| |
Collapse
|
17
|
Tonda-Turo C, Origlia N, Mattu C, Accorroni A, Chiono V. Current Limitations in the Treatment of Parkinson's and Alzheimer's Diseases: State-of-the-Art and Future Perspective of Polymeric Carriers. Curr Med Chem 2019; 25:5755-5771. [PMID: 29473493 DOI: 10.2174/0929867325666180221125759] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/18/2017] [Accepted: 02/06/2018] [Indexed: 12/18/2022]
Abstract
Alzheimer's and Parkinson's diseases are the most common neurodegenerative diseases worldwide and their incidence is increasing due to the aging population. At the moment, the available therapies are not disease modifying and have several limitations, some of which are discussed in this review. One of the main limitations of these treatments is the low concentration that drugs reach in the central nervous system after systemic administration. Indeed, the presence of biological barriers, particularly the blood-brain barrier (BBB), hinders the effective drug delivery to the brain, reducing the potential benefit coming from the administration of the medication. In this review, the mechanisms of transport across the BBB and new methods to improve drug passage across the BBB are discussed. These methods include non-invasive solutions such as intranasal and intravitreal administration, and the use of nanotechnology solutions based on polymeric carriers when the drug is intravenously injected, orally taken for intestine adsorption or delivered through the dermal mucosa. Also, it provides an analysis of more invasive solutions that include intracranially injected hydrogels and implanted devices for local drug delivery. Efforts in finding new therapeutic drugs blocking neurodegenerative disease progression or reverting their course should be coupled with efforts addressed to efficient drug delivery systems. Hence, new pharmacology discoveries together with advancements in nanotechnologies and biomaterials for regenerative medicine are required to effectively counteract neurodegenerative diseases.
Collapse
Affiliation(s)
- Chiara Tonda-Turo
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Turin, Italy
| | - Nicola Origlia
- CNR, Neuroscience Institute Via G. Moruzzi 1, 56124 Pisa, Italy
| | - Clara Mattu
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Turin, Italy
| | - Alice Accorroni
- CNR, Neuroscience Institute Via G. Moruzzi 1, 56124 Pisa, Italy.,Institute of Life Sciences, Scuola Superiore Sant'Anna, 56127 Pisa, Italy
| | - Valeria Chiono
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, 10129 Turin, Italy
| |
Collapse
|
18
|
Emerich DF, Kordower JH, Chu Y, Thanos C, Bintz B, Paolone G, Wahlberg LU. Widespread Striatal Delivery of GDNF from Encapsulated Cells Prevents the Anatomical and Functional Consequences of Excitotoxicity. Neural Plast 2019; 2019:6286197. [PMID: 30984255 PMCID: PMC6432730 DOI: 10.1155/2019/6286197] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Accepted: 02/11/2019] [Indexed: 12/14/2022] Open
Abstract
Methods Human ARPE-19 cells engineered to secrete high levels of the glial cell line-derived neurotrophic factor (GDNF) were encapsulated into hollow fiber membranes. The devices were implanted into the rat striatum 1 week prior to striatal quinolinic acid injections. Animals were evaluated using a battery of validated motor tests, and histology was performed to determine the extent of GDNF diffusion and associated prevention of neuronal cell loss and behavioral deficits. Results Encapsulated cell-based delivery of GDNF produced widespread distribution of GDNF throughout the entire implanted striatum. Stereological estimates of striatal neuron number and volume of lesion size revealed that GDNF delivery resulted in near complete neuroprotection. Conclusions Delivery of neurotrophic molecules such as GDNF using encapsulated cells has reached a technological point where clinical evaluation is justified. Because GDNF has been effective in animal models of Parkinson's disease, stroke, epilepsy, and Huntington's disease, among other debilitating neurodegenerative diseases, encapsulated cell-based delivery of GDNF might represent one innovative means of slowing the neural degeneration seen in a myriad of currently untreatable neurological diseases.
Collapse
Affiliation(s)
| | - Jeffrey H. Kordower
- Department of Neurological Sciences, Rush University Medical Center, Chicago Illinois, USA
| | - Yaping Chu
- Department of Neurological Sciences, Rush University Medical Center, Chicago Illinois, USA
| | | | | | - Giovanna Paolone
- Department of Diagnostic and Public Health, Section of Pharmacology, University of Verona P.le, LA Scuro, Verona, Italy
| | | |
Collapse
|
19
|
Galli E, Lindholm P, Kontturi LS, Saarma M, Urtti A, Yliperttula M. Characterization of CDNF-Secreting ARPE-19 Cell Clones for Encapsulated Cell Therapy. Cell Transplant 2019; 28:413-424. [PMID: 30841717 PMCID: PMC6628564 DOI: 10.1177/0963689719827943] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Cerebral dopamine neurotrophic factor (CDNF) shows beneficial effects in rodent models of Parkinson’s and Alzheimer’s disease. The brain is a challenging target for protein therapy due to its exclusive blood–brain barrier. Hence, the therapeutic protein should be delivered directly to the brain parenchyma. Implantation of encapsulated mammalian cells that constantly secrete CDNF is a potential approach for targeted and long-term protein delivery to the brain. In this study, we generated several CDNF-secreting cell clones derived from human retinal pigment epithelial cell line ARPE-19, and studied CDNF secretion from the clones maintained as monolayers and in polymeric microcapsules. The secretion of wild type (wt) CDNF transgene was low and the majority of the produced protein remained intracellular, locating mainly to the endoplasmic reticulum (ER). The secretion of wtCDNF decreased to even lower levels when the clones were in a non-dividing state, as in the microcapsules. Both codon optimization and deletion of the putative ER-retrieval signal (four last amino acids: KTEL) improved CDNF secretion. More importantly, the secretion of KTEL-deleted CDNF remained constant in the non-dividing clones. Thus, cells expressing KTEL-deleted CDNF, in contrast to wtCDNF, can be considered for cell encapsulation applications if the KTEL-deleted CDNF is proven to be biologically active in vivo.
Collapse
Affiliation(s)
- Emilia Galli
- 1 Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,2 Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Päivi Lindholm
- 2 Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Leena-Stiina Kontturi
- 1 Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- 2 Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Arto Urtti
- 1 Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,3 School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Marjo Yliperttula
- 1 Division of Pharmaceutical Biosciences, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| |
Collapse
|
20
|
Mitra S, Behbahani H, Eriksdotter M. Innovative Therapy for Alzheimer's Disease-With Focus on Biodelivery of NGF. Front Neurosci 2019; 13:38. [PMID: 30804738 PMCID: PMC6370742 DOI: 10.3389/fnins.2019.00038] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/15/2019] [Indexed: 12/31/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder associated with abnormal protein modification, inflammation and memory impairment. Aggregated amyloid beta (Aβ) and phosphorylated tau proteins are medical diagnostic features. Loss of memory in AD has been associated with central cholinergic dysfunction in basal forebrain, from where the cholinergic circuitry projects to cerebral cortex and hippocampus. Various reports link AD progression with declining activity of cholinergic neurons in basal forebrain. The neurotrophic molecule, nerve growth factor (NGF), plays a major role in the maintenance of cholinergic neurons integrity and function, both during development and adulthood. Numerous studies have also shown that NGF contributes to the survival and regeneration of neurons during aging and in age-related diseases such as AD. Changes in neurotrophic signaling pathways are involved in the aging process and contribute to cholinergic and cognitive decline as observed in AD. Further, gradual dysregulation of neurotrophic factors like NGF and brain derived neurotrophic factor (BDNF) have been reported during AD development thus intensifying further research in targeting these factors as disease modifying therapies against AD. Today, there is no cure available for AD and the effects of the symptomatic treatment like cholinesterase inhibitors (ChEIs) and memantine are transient and moderate. Although many AD treatment studies are being carried out, there has not been any breakthrough and new therapies are thus highly needed. Long-term effective therapy for alleviating cognitive impairment is a major unmet need. Discussion and summarizing the new advancements of using NGF as a potential therapeutic implication in AD are important. In summary, the intent of this review is describing available experimental and clinical data related to AD therapy, priming to gain additional facts associated with the importance of NGF for AD treatment, and encapsulated cell biodelivery (ECB) as an efficient tool for NGF delivery.
Collapse
Affiliation(s)
- Sumonto Mitra
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden
| | - Homira Behbahani
- Division of Neurogeriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Solna, Sweden
| | - Maria Eriksdotter
- Division of Clinical Geriatrics, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Huddinge, Sweden.,Aging Theme, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
21
|
Long-Term, Targeted Delivery of GDNF from Encapsulated Cells Is Neuroprotective and Reduces Seizures in the Pilocarpine Model of Epilepsy. J Neurosci 2019; 39:2144-2156. [PMID: 30665947 DOI: 10.1523/jneurosci.0435-18.2018] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 11/14/2018] [Accepted: 12/14/2018] [Indexed: 12/19/2022] Open
Abstract
Neurotrophic factors are candidates for treating epilepsy, but their development has been hampered by difficulties in achieving stable and targeted delivery of efficacious concentrations within the desired brain region. We have developed an encapsulated cell technology that overcomes these obstacles by providing a targeted, continuous, de novo synthesized source of high levels of neurotrophic molecules from human clonal ARPE-19 cells encapsulated into hollow fiber membranes. Here we illustrate the potential of this approach for delivering glial cell line-derived neurotrophic factor (GDNF) directly to the hippocampus of epileptic rats. In vivo studies demonstrated that bilateral intrahippocampal implants continued to secrete GDNF that produced high hippocampal GDNF tissue levels in a long-term manner. Identical implants robustly reduced seizure frequency in the pilocarpine model. Seizures were reduced rapidly, and this effect increased in magnitude over 3 months, ultimately leading to a reduction of seizures by 93%. This effect persisted even after device removal, suggesting potential disease-modifying benefits. Importantly, seizure reduction was associated with normalized changes in anxiety and improved cognitive performance. Immunohistochemical analyses revealed that the neurological benefits of GDNF were associated with the normalization of anatomical alterations accompanying chronic epilepsy, including hippocampal atrophy, cell degeneration, loss of parvalbumin-positive interneurons, and abnormal neurogenesis. These effects were associated with the activation of GDNF receptors. All in all, these results support the concept that the implantation of encapsulated GDNF-secreting cells can deliver GDNF in a sustained, targeted, and efficacious manner, paving the way for continuing preclinical evaluation and eventual clinical translation of this approach for epilepsy.SIGNIFICANCE STATEMENT Epilepsy is one of the most common neurological conditions, affecting millions of individuals of all ages. These patients experience debilitating seizures that frequently increase over time and can associate with significant cognitive decline and psychiatric disorders that are generally poorly controlled by pharmacotherapy. We have developed a clinically validated, implantable cell encapsulation system that delivers high and consistent levels of GDNF directly to the brain. In epileptic animals, this system produced a progressive and permanent reduction (>90%) in seizure frequency. These benefits were accompanied by improvements in cognitive and anxiolytic behavior and the normalization of changes in CNS anatomy that underlie chronic epilepsy. Together, these data suggest a novel means of tackling the frequently intractable neurological consequences of this devastating disorder.
Collapse
|
22
|
Farina M, Alexander JF, Thekkedath U, Ferrari M, Grattoni A. Cell encapsulation: Overcoming barriers in cell transplantation in diabetes and beyond. Adv Drug Deliv Rev 2019; 139:92-115. [PMID: 29719210 DOI: 10.1016/j.addr.2018.04.018] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/19/2018] [Accepted: 04/25/2018] [Indexed: 02/07/2023]
Abstract
Cell-based therapy is emerging as a promising strategy for treating a wide range of human diseases, such as diabetes, blood disorders, acute liver failure, spinal cord injury, and several types of cancer. Pancreatic islets, blood cells, hepatocytes, and stem cells are among the many cell types currently used for this strategy. The encapsulation of these "therapeutic" cells is under intense investigation to not only prevent immune rejection but also provide a controlled and supportive environment so they can function effectively. Some of the advanced encapsulation systems provide active agents to the cells and enable a complete retrieval of the graft in the case of an adverse body reaction. Here, we review various encapsulation strategies developed in academic and industrial settings, including the state-of-the-art technologies in advanced preclinical phases as well as those undergoing clinical trials, and assess their advantages and challenges. We also emphasize the importance of stimulus-responsive encapsulated cell systems that provide a "smart and live" therapeutic delivery to overcome barriers in cell transplantation as well as their use in patients.
Collapse
|
23
|
Nanobashvili A, Melin E, Emerich D, Tornøe J, Simonato M, Wahlberg L, Kokaia M. Unilateral ex vivo gene therapy by GDNF in epileptic rats. Gene Ther 2018; 26:65-74. [PMID: 30464254 DOI: 10.1038/s41434-018-0050-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 10/12/2018] [Accepted: 10/23/2018] [Indexed: 01/01/2023]
Abstract
Temporal lobe epilepsy (TLE) is the most common type of epilepsy in adults. This neurological disorder is characterized by focal seizures originating in the temporal lobe, often with secondary generalization. A variety of pharmacological treatments exist for patients suffering from focal seizures, but systemically administered drugs offer only symptomatic relief and frequently cause unwanted side effects. Moreover, available drugs are ineffective in one third of the epilepsy patients. Thus, developing more targeted and effective treatment strategies for focal seizures, originating from, e.g., the temporal lobe, is highly warranted. In order to deliver potential anti-epileptic agents directly into the seizure focus we used encapsulated cell biodelivery (ECB), a specific type of ex vivo gene therapy. Specifically, we asked whether unilateral delivery of glial cell line-derived neurotrophic factor (GDNF), exclusively into the epileptic focus, would suppress already established spontaneous recurrent seizures (SRS) in rats. Our results show that GDNF delivered by ECB devices unilaterally into the seizure focus in the hippocampus effectively decreases the number of SRS in epileptic rats. Thus, our study demonstrates that focal unilateral delivery of neurotrophic factors, such as GDNF, using ex vivo gene therapy based on ECB devices could be an effective anti-epileptic strategy providing a bases for the development of a novel, alternative, treatment for focal epilepsies.
Collapse
Affiliation(s)
| | - Esbjörn Melin
- Epilepsy Centre, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | | | | | - Michele Simonato
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Division of Neuroscience, University Vita-Salute San Raffaele, Milan, Italy
| | | | - Merab Kokaia
- Epilepsy Centre, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden.
| |
Collapse
|
24
|
Correia CR, Reis RL, Mano JF. Design Principles and Multifunctionality in Cell Encapsulation Systems for Tissue Regeneration. Adv Healthc Mater 2018; 7:e1701444. [PMID: 30102458 DOI: 10.1002/adhm.201701444] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 07/16/2018] [Indexed: 12/12/2022]
Abstract
Cell encapsulation systems are being increasingly applied as multifunctional strategies to regenerate tissues. Lessons afforded with encapsulation systems aiming to treat endocrine diseases seem to be highly valuable for the tissue engineering and regenerative medicine (TERM) systems of today, in which tissue regeneration and biomaterial integration are key components. Innumerous multifunctional systems for cell compartmentalization are being proposed to meet the specific needs required in the TERM field. Herein is reviewed the variable geometries proposed to produce cell encapsulation strategies toward tissue regeneration, including spherical and fiber-shaped systems, and other complex shapes and arrangements that better mimic the highly hierarchical organization of native tissues. The application of such principles in the TERM field brings new possibilities for the development of highly complex systems, which holds tremendous promise for tissue regeneration. The complex systems aim to recreate adequate environmental signals found in native tissue (in particular during the regenerative process) to control the cellular outcome, and conferring multifunctional properties, namely the incorporation of bioactive molecules and the ability to create smart and adaptative systems in response to different stimuli. The new multifunctional properties of such systems that are being employed to fulfill the requirements of the TERM field are also discussed.
Collapse
Affiliation(s)
- Clara R. Correia
- 3B's Research Group – Biomaterials, Biodegradables, and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark 4805‐017 Barco Guimarães Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| | - Rui L. Reis
- 3B's Research Group – Biomaterials, Biodegradables, and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark 4805‐017 Barco Guimarães Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| | - João F. Mano
- 3B's Research Group – Biomaterials, Biodegradables, and BiomimeticsUniversity of MinhoHeadquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine AvePark 4805‐017 Barco Guimarães Portugal
- ICVS/3B's – PT Government Associate Laboratory Braga/Guimarães Portugal
| |
Collapse
|
25
|
Boss C, Bouche N, De Marchi U. Encapsulated Optically Responsive Cell Systems: Toward Smart Implants in Biomedicine. Adv Healthc Mater 2018; 7:e1701148. [PMID: 29283209 DOI: 10.1002/adhm.201701148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 11/06/2017] [Indexed: 01/09/2023]
Abstract
Managing increasingly prevalent chronic diseases will require close continuous monitoring of patients. Cell-based biosensors may be used for implantable diagnostic systems to monitor health status. Cells are indeed natural sensors in the body. Functional cellular systems can be maintained in the body for long-term implantation using cell encapsulation technology. By taking advantage of recent progress in miniaturized optoelectronic systems, the genetic engineering of optically responsive cells may be combined with cell encapsulation to generate smart implantable cell-based sensing systems. In biomedical research, cell-based biosensors may be used to study cell signaling, therapeutic effects, and dosing of bioactive molecules in preclinical models. Today, a wide variety of genetically encoded fluorescent sensors have been developed for real-time imaging of living cells. Here, recent developments in genetically encoded sensors, cell encapsulation, and ultrasmall optical systems are highlighted. The integration of these components in a new generation of biosensors is creating innovative smart in vivo cell-based systems, bringing novel perspectives for biomedical research and ultimately allowing unique health monitoring applications.
Collapse
Affiliation(s)
- Christophe Boss
- Device EngineeringNestlé Institute of Health Sciences EPFL Innovation Park Lausanne CH‐1015 Switzerland
| | - Nicolas Bouche
- Device EngineeringNestlé Institute of Health Sciences EPFL Innovation Park Lausanne CH‐1015 Switzerland
| | - Umberto De Marchi
- Mitochondrial FunctionNestlé Institute of Health Sciences EPFL Innovation Park Lausanne CH‐1015 Switzerland
| |
Collapse
|
26
|
Gonzalez-Pujana A, Orive G, Pedraz JL, Santos-Vizcaino E, Hernandez RM. Alginate Microcapsules for Drug Delivery. SPRINGER SERIES IN BIOMATERIALS SCIENCE AND ENGINEERING 2018. [DOI: 10.1007/978-981-10-6910-9_3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
27
|
Designing a retrievable and scalable cell encapsulation device for potential treatment of type 1 diabetes. Proc Natl Acad Sci U S A 2017; 115:E263-E272. [PMID: 29279393 DOI: 10.1073/pnas.1708806115] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Cell encapsulation has been shown to hold promise for effective, long-term treatment of type 1 diabetes (T1D). However, challenges remain for its clinical applications. For example, there is an unmet need for an encapsulation system that is capable of delivering sufficient cell mass while still allowing convenient retrieval or replacement. Here, we report a simple cell encapsulation design that is readily scalable and conveniently retrievable. The key to this design was to engineer a highly wettable, Ca2+-releasing nanoporous polymer thread that promoted uniform in situ cross-linking and strong adhesion of a thin layer of alginate hydrogel around the thread. The device provided immunoprotection of rat islets in immunocompetent C57BL/6 mice in a short-term (1-mo) study, similar to neat alginate fibers. However, the mechanical property of the device, critical for handling and retrieval, was much more robust than the neat alginate fibers due to the reinforcement of the central thread. It also had facile mass transfer due to the short diffusion distance. We demonstrated the therapeutic potential of the device through the correction of chemically induced diabetes in C57BL/6 mice using rat islets for 3 mo as well as in immunodeficient SCID-Beige mice using human islets for 4 mo. We further showed, as a proof of concept, the scalability and retrievability in dogs. After 1 mo of implantation in dogs, the device could be rapidly retrieved through a minimally invasive laparoscopic procedure. This encapsulation device may contribute to a cellular therapy for T1D because of its retrievability and scale-up potential.
Collapse
|
28
|
Fransson A, Tornøe J, Wahlberg LU, Ulfendahl M. The feasibility of an encapsulated cell approach in an animal deafness model. J Control Release 2017; 270:275-281. [PMID: 29269144 PMCID: PMC5819869 DOI: 10.1016/j.jconrel.2017.12.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/15/2017] [Accepted: 12/17/2017] [Indexed: 01/18/2023]
Abstract
For patients with profound hearing loss a cochlear implant (CI) is the only treatment today. The function of a CI depends in part of the function and survival of the remaining spiral ganglion neurons (SGN). It is well known from animal models that inner ear infusion of neurotrophic factors prevents SGN degeneration and maintains electrical responsiveness in deafened animals. The purpose with this study was to investigate the effects of a novel encapsulated cell (EC) device releasing neurotrophic factors in the deafened guinea pig. The results showed that an EC device releasing glial cell line-derived neurotrophic factor (GDNF) or brain-derived neurotrophic factor (BDNF) implanted for four weeks in deafened guinea pigs significantly preserved the SGNs and maintained their electrical responsiveness. There was a significant difference between BDNF and GDNF in favour of GDNF. This study, demonstrating positive structural and functional effects in the deafened inner ear, suggests that an implanted EC device releasing biologically protective substances offers a feasible approach for treating progressive hearing impairment.
Collapse
Affiliation(s)
- Anette Fransson
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | | | | | - Mats Ulfendahl
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
29
|
Gonzalez-Pujana A, Santos E, Orive G, Pedraz JL, Hernandez RM. Cell microencapsulation technology: Current vision of its therapeutic potential through the administration routes. J Drug Deliv Sci Technol 2017. [DOI: 10.1016/j.jddst.2017.03.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
30
|
Combining NT3-overexpressing MSCs and PLGA microcarriers for brain tissue engineering: A potential tool for treatment of Parkinson's disease. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 76:934-943. [PMID: 28482609 DOI: 10.1016/j.msec.2017.02.178] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 01/03/2017] [Accepted: 02/28/2017] [Indexed: 12/18/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that characterized by destruction of substantia nigrostriatal pathway due to the loss of dopaminergic (DA) neurons. Regardless of substantial efforts for treatment of PD in recent years, an effective therapeutic strategy is still missing. In a multidisciplinary approach, bone marrow derived mesenchymal stem cells (BMSCs) are genetically engineered to overexpress neurotrophin-3 (nt-3 gene) that protect central nervous system tissues and stimulates neuronal-like differentiation of BMSCs. Poly(lactic-co-glycolic acid) (PLGA) microcarriers are designed as an injectable scaffold and synthesized via double emulsion method. The surface of PLGA microcarriers are functionalized by collagen as a bioadhesive agent for improved cell attachment. The results demonstrate effective overexpression of NT-3. The expression of tyrosine hydroxylase (TH) in transfected BMSCs reveal that NT-3 promotes the intracellular signaling pathway of DA neuron differentiation. It is also shown that transfected BMSCs are successfully attached to the surface of microcarriers. The presence of dopamine in peripheral media of cell/microcarrier complex reveals that BMSCs are successfully differentiated into dopaminergic neuron. Our approach that sustains presence of growth factor can be suggested as a novel complementary therapeutic strategy for treatment of Parkinson disease.
Collapse
|
31
|
Kimura A, Namekata K, Guo X, Harada C, Harada T. Neuroprotection, Growth Factors and BDNF-TrkB Signalling in Retinal Degeneration. Int J Mol Sci 2016; 17:ijms17091584. [PMID: 27657046 PMCID: PMC5037849 DOI: 10.3390/ijms17091584] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 09/01/2016] [Accepted: 09/14/2016] [Indexed: 12/18/2022] Open
Abstract
Neurotrophic factors play key roles in the development and survival of neurons. The potent neuroprotective effects of neurotrophic factors, including brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF), glial cell-line derived neurotrophic factor (GDNF) and nerve growth factor (NGF), suggest that they are good therapeutic candidates for neurodegenerative diseases. Glaucoma is a neurodegenerative disease of the eye that causes irreversible blindness. It is characterized by damage to the optic nerve, usually due to high intraocular pressure (IOP), and progressive degeneration of retinal neurons called retinal ganglion cells (RGCs). Current therapy for glaucoma focuses on reduction of IOP, but neuroprotection may also be beneficial. BDNF is a powerful neuroprotective agent especially for RGCs. Exogenous application of BDNF to the retina and increased BDNF expression in retinal neurons using viral vector systems are both effective in protecting RGCs from damage. Furthermore, induction of BDNF expression by agents such as valproic acid has also been beneficial in promoting RGC survival. In this review, we discuss the therapeutic potential of neurotrophic factors in retinal diseases and focus on the differential roles of glial and neuronal TrkB in neuroprotection. We also discuss the role of neurotrophic factors in neuroregeneration.
Collapse
Affiliation(s)
- Atsuko Kimura
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.
| | - Kazuhiko Namekata
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.
| | - Xiaoli Guo
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.
| | - Chikako Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.
| | - Takayuki Harada
- Visual Research Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan.
| |
Collapse
|
32
|
Åkesson E, Sundström E. Human neural progenitor cells in central nervous system lesions. Best Pract Res Clin Obstet Gynaecol 2015; 31:69-81. [PMID: 26803559 DOI: 10.1016/j.bpobgyn.2015.11.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 11/30/2015] [Indexed: 12/13/2022]
Abstract
Various immature cells can be isolated from human embryonic and fetal central nervous system (CNS) residual tissue and potentially be used in cell therapy for a number of neurological diseases and CNS insults. Transplantation of neural stem and progenitor cells is essential for replacing lost cells, particularly in the CNS with very limited endogenous regenerative capacity. However, while dopamine released from transplanted cells can substitute the lost dopamine neurons in the experimental models of Parkinson's disease, stem and progenitor cells primarily have a neuroprotective effect, probably through the release of trophic factors. Understanding the therapeutic effects of transplanted cells is crucial to determine the design of clinical trials. During the last few years, a number of clinical trials for CNS diseases and insults such as amyotrophic lateral sclerosis (ALS), stroke, and spinal cord trauma using neural progenitor cells have been initiated. Data from these early studies will provide vital information on the safety of transplanting these cells, which still is a major concern. That the beneficial results observed in experimental models also can be repeated in the clinical setting is highly hoped for.
Collapse
Affiliation(s)
- Elisabet Åkesson
- Division of Neurodegeneration, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Novum 5th Floor, S-14157, Huddinge, and Stockholm Sjukhem Foundation, Box 12230, S-10226 Stockholm, Sweden
| | - Erik Sundström
- Division of Neurodegeneration, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Novum 5th Floor, S-14157, Huddinge, and Stockholm Sjukhem Foundation, Box 12230, S-10226 Stockholm, Sweden.
| |
Collapse
|
33
|
Hashemi M, Kalalinia F. Application of encapsulation technology in stem cell therapy. Life Sci 2015; 143:139-46. [DOI: 10.1016/j.lfs.2015.11.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Revised: 10/15/2015] [Accepted: 11/06/2015] [Indexed: 11/26/2022]
|
34
|
Current Neurogenic and Neuroprotective Strategies to Prevent and Treat Neurodegenerative and Neuropsychiatric Disorders. Neuromolecular Med 2015; 17:404-22. [PMID: 26374113 DOI: 10.1007/s12017-015-8369-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 08/22/2015] [Indexed: 12/31/2022]
Abstract
The adult central nervous system is commonly known to have a very limited regenerative capacity. The presence of functional stem cells in the brain can therefore be seen as a paradox, since in other organs these are known to counterbalance cell loss derived from pathological conditions. This fact has therefore raised the possibility to stimulate neural stem cell differentiation and proliferation or survival by either stem cell replacement therapy or direct administration of neurotrophic factors or other proneurogenic molecules, which in turn has also originated regenerative medicine for the treatment of otherwise incurable neurodegenerative and neuropsychiatric disorders that take a huge toll on society. This may be facilitated by the fact that many of these disorders converge on similar pathophysiological pathways: excitotoxicity, oxidative stress, neuroinflammation, mitochondrial failure, excessive intracellular calcium and apoptosis. This review will therefore focus on the most promising achievements in promoting neuroprotection and neuroregeneration reported to date.
Collapse
|
35
|
Merighi A. Targeting the glial-derived neurotrophic factor and related molecules for controlling normal and pathologic pain. Expert Opin Ther Targets 2015; 20:193-208. [PMID: 26863504 DOI: 10.1517/14728222.2016.1085972] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
INTRODUCTION Glial-derived neurotrophic factor (GDNF) and its family of ligands (GFLs) have several functions in the nervous system. As a survival factor for dopaminergic neurons, GDNF was used in clinical trials for Parkinson's disease. GFLs and their receptors are also potential targets for new pain-controlling drugs. Although molecules with analgesic activities in rodents mostly failed to be effective in translational studies, this potential should not be underestimated. AREAS COVERED The circuitry, molecular, and cellular mechanisms by which GFLs control nociception and their intervention in inflammatory and neuropathic pain are considered first. The problems related to effective GDNF delivery to the brain and the possibility to target the GFL receptor complex rather than its ligands are then discussed, also considering the use of non-peptidyl agonists. EXPERT OPINION In nociceptive pathways, an ideal drug should either: i) target the release of endogenous GFLs from large dense-cored vesicles (LGVs) by acting, for example, onto the phosphatidylinositol-3-phosphate [PtdIns(3)P] pool, which is sensitive to Ca(2+) modulation, or ii) target the GFL receptor complex. Besides XIB403, a tiol molecule that enhances GFRα family receptor signaling, existing drugs such as retinoic acid and amitriptyline should be considered for effective targeting of GDNF, at least in neuropathic pain. The approach of pain modeling in experimental animals is discussed.
Collapse
Affiliation(s)
- Adalberto Merighi
- a University of Turin, Department of Veterinary Sciences , Grugliasco, TO, Italy ;
| |
Collapse
|
36
|
Cell encapsulation: technical and clinical advances. Trends Pharmacol Sci 2015; 36:537-46. [DOI: 10.1016/j.tips.2015.05.003] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Revised: 05/13/2015] [Accepted: 05/14/2015] [Indexed: 01/18/2023]
|
37
|
|
38
|
d'Anglemont de Tassigny X, Pascual A, López-Barneo J. GDNF-based therapies, GDNF-producing interneurons, and trophic support of the dopaminergic nigrostriatal pathway. Implications for Parkinson's disease. Front Neuroanat 2015; 9:10. [PMID: 25762899 PMCID: PMC4327623 DOI: 10.3389/fnana.2015.00010] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 01/21/2015] [Indexed: 01/09/2023] Open
Abstract
The glial cell line-derived neurotrophic factor (GDNF) is a well-established trophic agent for dopaminergic (DA) neurons in vitro and in vivo. GDNF is necessary for maintenance of neuronal morphological and neurochemical phenotype and protects DA neurons from toxic damage. Numerous studies on animal models of Parkinson’s disease (PD) have reported beneficial effects of GDNF on nigrostriatal DA neuron survival. However, translation of these observations to the clinical setting has been hampered so far by side effects associated with the chronic continuous intra-striatal infusion of recombinant GDNF. In addition, double blind and placebo-controlled clinical trials have not reported any clinically relevant effect of GDNF on PD patients. In the past few years, experiments with conditional Gdnf knockout mice have suggested that GDNF is necessary for maintenance of DA neurons in adulthood. In parallel, new methodologies for exogenous GDNF delivery have been developed. Recently, it has been shown that a small population of scattered, electrically interconnected, parvalbumin positive (PV+) GABAergic interneurons is responsible for most of the GDNF produced in the rodent striatum. In addition, cholinergic striatal interneurons appear to be also involved in the modulation of striatal GDNF. In this review, we summarize current knowledge on brain GDNF delivery, homeostasis, and its effects on nigrostriatal DA neurons. Special attention is paid to the therapeutic potential of endogenous GDNF stimulation in PD.
Collapse
Affiliation(s)
- Xavier d'Anglemont de Tassigny
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville, Spain
| | - Alberto Pascual
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville, Spain
| | - José López-Barneo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville, Spain ; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla Seville, Spain ; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) Madrid, Spain
| |
Collapse
|
39
|
Coatti GC, Beccari MS, Olávio TR, Mitne-Neto M, Okamoto OK, Zatz M. Stem cells for amyotrophic lateral sclerosis modeling and therapy: Myth or fact? Cytometry A 2015; 87:197-211. [DOI: 10.1002/cyto.a.22630] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 12/28/2014] [Indexed: 02/06/2023]
Affiliation(s)
- G. C. Coatti
- Human Genome and Stem Cell Research Center; Department of Genetics and Evolutionary Biology, Biosciences Institute, University of São Paulo (USP); São Paulo Brazil
| | - M. S. Beccari
- Human Genome and Stem Cell Research Center; Department of Genetics and Evolutionary Biology, Biosciences Institute, University of São Paulo (USP); São Paulo Brazil
| | - T. R. Olávio
- Human Genome and Stem Cell Research Center; Department of Genetics and Evolutionary Biology, Biosciences Institute, University of São Paulo (USP); São Paulo Brazil
| | - M. Mitne-Neto
- Human Genome and Stem Cell Research Center; Department of Genetics and Evolutionary Biology, Biosciences Institute, University of São Paulo (USP); São Paulo Brazil
- Fleury Group (Research and Development Department); São Paulo Brazil
| | - O. K. Okamoto
- Human Genome and Stem Cell Research Center; Department of Genetics and Evolutionary Biology, Biosciences Institute, University of São Paulo (USP); São Paulo Brazil
| | - M. Zatz
- Human Genome and Stem Cell Research Center; Department of Genetics and Evolutionary Biology, Biosciences Institute, University of São Paulo (USP); São Paulo Brazil
| |
Collapse
|
40
|
Pienaar IS, Lee CH, Elson JL, McGuinness L, Gentleman SM, Kalaria RN, Dexter DT. Deep-brain stimulation associates with improved microvascular integrity in the subthalamic nucleus in Parkinson's disease. Neurobiol Dis 2014; 74:392-405. [PMID: 25533682 DOI: 10.1016/j.nbd.2014.12.006] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 12/01/2014] [Accepted: 12/05/2014] [Indexed: 12/25/2022] Open
Abstract
Deep brain stimulation (DBS) of the subthalamic nucleus (STN) has become an accepted treatment for motor symptoms in a subset of Parkinson's disease (PD) patients. The mechanisms why DBS is effective are incompletely understood, but previous studies show that DBS targeted in brain structures other than the STN may modify the microvasculature. However, this has not been studied in PD subjects who have received STN-DBS. Here we investigated the extent and nature of microvascular changes in post-mortem STN samples from STN-DBS PD patients, compared to aged controls and PD patients who had not been treated with STN-DBS. We used immunohistochemical and immunofluorescent methods to assess serial STN-containing brain sections from PD and STN-DBS PD cases, compared to similar age controls using specific antibodies to detect capillaries, an adherens junction and tight junction-associated proteins as well as activated microglia. Cellular features in stained sections were quantified by confocal fluorescence microscopy and stereological methods in conjunction with in vitro imaging tools. We found significant upregulation of microvessel endothelial cell thickness, length and density but lowered activated microglia density and striking upregulation of all analysed adherens junction and tight junction-associated proteins in STN-DBS PD patients compared to non-DBS PD patients and controls. Moreover, in STN-DBS PD samples, expression of an angiogenic factor, vascular endothelial growth factor (VEGF), was significantly upregulated compared to the other groups. Our findings suggest that overexpressed VEGF and downregulation of inflammatory processes may be critical mechanisms underlying the DBS-induced microvascular changes.
Collapse
Affiliation(s)
- Ilse S Pienaar
- Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, United Kingdom.
| | - Cecilia Heyne Lee
- The Sir William Dunn School of Pathology, South Parks Road, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Joanna L Elson
- Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne NE1 3BZ, United Kingdom; Centre for Human Metabonomics, North-West University, Potchefstroom, South Africa
| | - Louisa McGuinness
- Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Stephen M Gentleman
- Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, United Kingdom
| | - Raj N Kalaria
- Institute of Neuroscience, Newcastle University, Campus for Ageing and Vitality, Newcastle upon Tyne NE4 5PL, United Kingdom
| | - David T Dexter
- Centre for Neuroinflammation & Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, United Kingdom
| |
Collapse
|
41
|
Herrán E, Requejo C, Ruiz-Ortega JA, Aristieta A, Igartua M, Bengoetxea H, Ugedo L, Pedraz JL, Lafuente JV, Hernández RM. Increased antiparkinson efficacy of the combined administration of VEGF- and GDNF-loaded nanospheres in a partial lesion model of Parkinson's disease. Int J Nanomedicine 2014; 9:2677-87. [PMID: 24920904 PMCID: PMC4043720 DOI: 10.2147/ijn.s61940] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Current research efforts are focused on the application of growth factors, such as glial cell line-derived neurotrophic factor (GDNF) and vascular endothelial growth factor (VEGF), as neuroregenerative approaches that will prevent the neurodegenerative process in Parkinson’s disease. Continuing a previous work published by our research group, and with the aim to overcome different limitations related to growth factor administration, VEGF and GDNF were encapsulated in poly(lactic-co-glycolic acid) nanospheres (NS). This strategy facilitates the combined administration of the VEGF and GDNF into the brain of 6-hydroxydopamine (6-OHDA) partially lesioned rats, resulting in a continuous and simultaneous drug release. The NS particle size was about 200 nm and the simultaneous addition of VEGF NS and GDNF NS resulted in significant protection of the PC-12 cell line against 6-OHDA in vitro. Once the poly(lactic-co-glycolic acid) NS were implanted into the striatum of 6-OHDA partially lesioned rats, the amphetamine rotation behavior test was carried out over 10 weeks, in order to check for in vivo efficacy. The results showed that VEGF NS and GDNF NS significantly decreased the number of amphetamine-induced rotations at the end of the study. In addition, tyrosine hydroxylase immunohistochemical analysis in the striatum and the external substantia nigra confirmed a significant enhancement of neurons in the VEGF NS and GDNF NS treatment group. The synergistic effect of VEGF NS and GDNF NS allows for a reduction of the dose by half, and may be a valuable neurogenerative/neuroreparative approach for treating Parkinson’s disease.
Collapse
Affiliation(s)
- Enara Herrán
- NanoBioCel Group, Laboratory of Pharmaceutics, University of the Basque Country (UPV/EHU), School of Pharmacy, Vitoria, Spain ; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria, Spain
| | - Catalina Requejo
- LaNCE, Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
| | | | - Asier Aristieta
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Manoli Igartua
- NanoBioCel Group, Laboratory of Pharmaceutics, University of the Basque Country (UPV/EHU), School of Pharmacy, Vitoria, Spain ; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria, Spain
| | - Harkaitz Bengoetxea
- LaNCE, Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Luisa Ugedo
- Department of Pharmacology, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Jose Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, University of the Basque Country (UPV/EHU), School of Pharmacy, Vitoria, Spain ; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria, Spain
| | - Jose Vicente Lafuente
- LaNCE, Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Rosa Maria Hernández
- NanoBioCel Group, Laboratory of Pharmaceutics, University of the Basque Country (UPV/EHU), School of Pharmacy, Vitoria, Spain ; Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Vitoria, Spain
| |
Collapse
|
42
|
Lundkvist J, Halldin MM, Sandin J, Nordvall G, Forsell P, Svensson S, Jansson L, Johansson G, Winblad B, Ekstrand J. The battle of Alzheimer's Disease - the beginning of the future Unleashing the potential of academic discoveries. Front Pharmacol 2014; 5:102. [PMID: 24847271 PMCID: PMC4023025 DOI: 10.3389/fphar.2014.00102] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 04/18/2014] [Indexed: 01/21/2023] Open
Abstract
Alzheimer's Disease (AD) is the most common form of dementia, affecting approximately 36 million people worldwide. To date there is no preventive or curative treatment available for AD, and in absence of major progress in therapeutic development, AD manifests a concrete socioeconomic threat. The awareness of the growing problem of AD is increasing, exemplified by the recent G8 Dementia Summit, a meeting held in order to set the stage and steer the compass for the future. Simultaneously, and paradoxically, we have seen key players in the pharmaceutical industry that have recently closed or significantly decreased their R&D spending on AD and other CNS disorders. Given the pressing need for new treatments in this area, other actors need to step-in and enter this drug discovery arena complementing the industrial efforts, in order to turn biological and technological progress into novel therapeutics. In this article, we present an example of a novel drug discovery initiative that in a non-profit setting, aims to integrate with both preclinical and clinical academic groups and pharmaceutical industry to explore the therapeutic potential of new concepts in patients, using novel biology, state of the art technologies and rapid concept testing.
Collapse
Affiliation(s)
- Johan Lundkvist
- AlzeCure Foundation, Karolinska Institutet Science Park NovumHuddinge, Sweden
| | - Magnus M. Halldin
- AlzeCure Foundation, Karolinska Institutet Science Park NovumHuddinge, Sweden
| | - Johan Sandin
- AlzeCure Foundation, Karolinska Institutet Science Park NovumHuddinge, Sweden
| | - Gunnar Nordvall
- AlzeCure Foundation, Karolinska Institutet Science Park NovumHuddinge, Sweden
| | - Pontus Forsell
- AlzeCure Foundation, Karolinska Institutet Science Park NovumHuddinge, Sweden
| | - Samuel Svensson
- AlzeCure Foundation, Karolinska Institutet Science Park NovumHuddinge, Sweden
| | | | - Gunilla Johansson
- Center for Alzheimer Research at Karolinska Institutet and Swedish Brain PowerNovum, Huddinge, Sweden
| | - Bengt Winblad
- Center for Alzheimer Research at Karolinska Institutet and Swedish Brain PowerNovum, Huddinge, Sweden
| | - Jonas Ekstrand
- AlzeCure Foundation, Karolinska Institutet Science Park NovumHuddinge, Sweden
| |
Collapse
|
43
|
Emerich DF, Orive G, Thanos C, Tornoe J, Wahlberg LU. Encapsulated cell therapy for neurodegenerative diseases: from promise to product. Adv Drug Deliv Rev 2014; 67-68:131-41. [PMID: 23880505 DOI: 10.1016/j.addr.2013.07.008] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 05/31/2013] [Accepted: 07/12/2013] [Indexed: 12/27/2022]
Abstract
Delivering therapeutic molecules, including trophic factor proteins, across the blood brain barrier to the brain parenchyma to treat chronic neurodegenerative diseases remains one of the great challenges in biology. To be effective, delivery needs to occur in a long-term and stable manner at sufficient quantities directly to the target region in a manner that is selective but yet covers enough of the target site to be efficacious. One promising approach uses cellular implants that produce and deliver therapeutic molecules directly to the brain region of interest. Implanted cells can be precisely positioned into the desired region and can be protected from host immunological attack by encapsulating them and by surrounding them within an immunoisolatory, semipermeable capsule. In this approach, cells are enclosed within a semiporous capsule with a perm selective membrane barrier that admits oxygen and required nutrients and releases bioactive cell secretions while restricting passage of larger cytotoxic agents from the host immune defense system. Recent advances in human cell line development have increased the levels of secreted therapeutic molecules from encapsulated cells, and membrane extrusion techniques have led to the first ever clinical demonstrations of long-term survival and function of encapsulated cells in the brain parenchyma. As such, cell encapsulation is capable of providing a targeted, continuous, de novo synthesized source of very high levels of therapeutic molecules that can be distributed over significant portions of the brain.
Collapse
|
44
|
Chiu WT, Lin CM, Tsai TC, Wu CW, Tsai CL, Lin SH, Chen JJJ. Real-time electrochemical recording of dopamine release under optogenetic stimulation. PLoS One 2014; 9:e89293. [PMID: 24586667 PMCID: PMC3930700 DOI: 10.1371/journal.pone.0089293] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Accepted: 01/20/2014] [Indexed: 01/02/2023] Open
Abstract
Dopaminergic PC12 cells can synthesize and release dopamine, providing a good cellular model for investigating dopamine regulation. Optogenetic stimulation of channelrhodopsin-2 provides high spatial and temporal precision for selective stimulation as a powerful neuromodulation tool for neuroscience studies. The aim of this study is to measure dopamine release from dopaminergic PC12 cells under optogenetic stimulation using electrochemical recording of self-assembled monolayers modified microelectrode with amperometric measurement in real time. The activation of PC12 cells under various optogenetic stimulation schemes are characterized by measuring single-cell Ca2+ imaging. After 10 seconds of optogenetic stimulation, the evoked intracellular Ca2+ level and dopamine current of channelrhodopsin-2-transfected PC12 cells were 1.6- and 3.5-fold higher than those of the control cells. The optogenetic stimulation effects on Ca2+ influx and dopamine release were 81% and 63% inhibition by using a Ca2+ channel antagonist Nifedipine. The results indicate that optogenetic stimulation can evoke voltage-gated Ca2+ channel-dependent dopamine exocytosis from PC12 cells in a cell specific, temporally precise and dose-dependent manner. This proposed dopamine recording system can be developed to be a good cell model for dopamine regulation and drug screening in vitro, or dopaminergic cell implantation therapy in vivo using optogenetic stimulation in a precise and convenient way.
Collapse
Affiliation(s)
- Wen-Tai Chiu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
- Institute of Basic Medical Sciences, National Cheng Kung University, Tainan, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
| | - Che-Ming Lin
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Tien-Chun Tsai
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Chun-Wei Wu
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Ching-Lin Tsai
- Department of Marine Biotechnology and Resources, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Sheng-Hsiang Lin
- Institute of Clinical Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jia-Jin Jason Chen
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, Taiwan
- National Applied Research Laboratories, Taipei, Taiwan
- * E-mail:
| |
Collapse
|
45
|
Huang SL, Wang J, He XJ, Li ZF, Pu JN, Shi W. Secretion of BDNF and GDNF from free and encapsulated choroid plexus epithelial cells. Neurosci Lett 2014; 566:42-5. [PMID: 24561094 DOI: 10.1016/j.neulet.2014.02.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 02/08/2014] [Accepted: 02/10/2014] [Indexed: 10/25/2022]
Abstract
Choroid plexus epithelial cells secrete numerous biologically active neurotrophic factors, which may be beneficial to the transplantation site. Encapsulated cells are often used in tissue transplantation. The present study was conducted to investigate the effect of encapsulation on the secretory function of choroid plexus epithelial cells. Neonatal rat choroid plexus epithelial cells were primarily cultured. After 9 days of culture, the cells were distributed into two groups, and one group of cells was encapsulated in vitro. The initial culture conditions such as cell numbers and medium volumes were the same. Supernatants in the free and encapsulated choroid plexus epithelial cells were collected at the time points of day 1 through day 7. Quantitative determination of the BDNF and GDNF levels was performed by enzyme-linked immunosorbent assay to assess the secretory function of the cells in the two forms. Statistical analyses were performed using a Student t test. P<0.05 was set to indicate statistical significance. A very similar secretion pattern was observed in both groups. In the first 4 days of encapsulation, the release of BDNF and GDNF in the encapsulated cells was significantly lower than that in the free cells, while the difference diminished after day 5. This in vitro study demonstrates that the secretion of BDNF and GDNF in encapsulated choroid plexus epithelial cells is different from that in non-encapsulated cells in the early stage of encapsulation treatment, whereas it is similar in the later stage.
Collapse
Affiliation(s)
- Sheng-Li Huang
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Jing Wang
- Department of pediatrics, Xi'an Children's Hospital, Xi'an, China
| | - Xi-Jing He
- Department of Orthopaedics, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Zong-Fang Li
- Central Laboratory for scientific Research, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Jing-Nan Pu
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Wei Shi
- Department of Neurosurgery, the Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
46
|
Sommer DB, Stacy MA. What’s in the pipeline for the treatment of Parkinson’s disease? Expert Rev Neurother 2014; 8:1829-39. [DOI: 10.1586/14737175.8.12.1829] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
47
|
α-Synuclein vaccination prevents the accumulation of parkinson disease-like pathologic inclusions in striatum in association with regulatory T cell recruitment in a rat model. J Neuropathol Exp Neurol 2013; 72:624-45. [PMID: 23771222 DOI: 10.1097/nen.0b013e31829768d2] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Human leukocyte antigen-DR induction and lymphocyte infiltrates in the brains of patients with Parkinson disease (PD) and the presence in serum of α-synuclein (α-syn)-specific antibodies suggest that the peripheral immune system may have an active role in the progression of PD. We designed a vaccination strategy to attempt to control these processes and mediate protection against disease progression in a rat PD model. Using a recombinant adeno-associated viral vector, we unilaterally overexpressed human α-syn in the rat substantia nigra to induce a progressive neuropathologic process. Prior to stereotactic delivery of the viral vector, animals were vaccinated with recombinant α-syn (asyn). This resulted in a high-titer anti-α-syn antibody response on α-syn overexpression; the accumulation of CD4-positive, MHC II-positive ramified microglia in the substantia nigra; long-lasting infiltration of CD4-positive, Foxp3-positive cells throughout the nigrostriatal system; and fewer pathologic aggregates in the striatum versus control animals that had received a mock vaccine. A long-term increase in GDNF levels in the striatum and IgG deposition in α-syn-overexpressing cells and neurites in the substantia nigra were also observed. Together, these results suggest that a protective vaccination strategy results in induction of regulatory T cells and distinctly activated microglia, and that this can induce immune tolerance against α-syn.
Collapse
|
48
|
Glavaski-Joksimovic A, Bohn MC. Mesenchymal stem cells and neuroregeneration in Parkinson's disease. Exp Neurol 2013; 247:25-38. [DOI: 10.1016/j.expneurol.2013.03.016] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 03/14/2013] [Indexed: 02/06/2023]
|
49
|
Kordower JH, Bjorklund A. Trophic factor gene therapy for Parkinson's disease. Mov Disord 2013; 28:96-109. [PMID: 23390096 DOI: 10.1002/mds.25344] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 12/05/2012] [Accepted: 12/13/2012] [Indexed: 11/06/2022] Open
Abstract
Parkinson's disease (PD) is a chronic and progressive neurodegenerative movement disorder for which there is presently no cure. Pharmacological remedies targeting the dopaminergic network are relatively effective at ameliorating motor deficits, especially in the early stages of the disease, but none of these therapies are curative and many generate their own problems. Recent advances in PD research have demonstrated that gene delivery of trophic factors, glial cell line-derived neurotrophic factor (GDNF) and neurturin, in particular, can provide structural and functional recovery in rodent and nonhuman primate models of PD. Similar success has been gleaned in open-label clinical trials, although this has yet to be realized in double-blinded analyses. This work reviews the field of trophic factor gene delivery for PD.
Collapse
Affiliation(s)
- Jeffrey H Kordower
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois 60612, USA.
| | | |
Collapse
|
50
|
Angelova A, Angelov B, Drechsler M, Lesieur S. Neurotrophin delivery using nanotechnology. Drug Discov Today 2013; 18:1263-71. [PMID: 23891881 DOI: 10.1016/j.drudis.2013.07.010] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 07/01/2013] [Accepted: 07/16/2013] [Indexed: 12/13/2022]
Abstract
Deficits or overexpression of neurotrophins cause neurodegenerative diseases and psychiatric disorders. These proteins are required for the maintenance of the function, plasticity and survival of neurons in the central (CNS) and peripheral nervous systems. Significant efforts have been devoted to developing therapeutic delivery systems that enable control of neurotrophin dosage in the brain. Here, we suggest that nanoparticulate carriers favoring targeted delivery in specific brain areas and minimizing biodistribution to the systemic circulation should be developed toward clinical benefits of neuroregeneration. We also provide examples of improved targeted neurotrophin delivery to localized areas in the CNS.
Collapse
Affiliation(s)
- Angelina Angelova
- CNRS UMR8612 Institut Galien Paris-Sud, 5 rue J.B. Clément, F-92296 Châtenay-Malabry cedex, France; University Paris Sud 11, Faculté de Pharmacie, LabEx LERMIT, Châtenay-Malabry, France.
| | | | | | | |
Collapse
|