1
|
Lin MT, Chan TY, Liao WH, Wu CH, Young TH, Chen WS. Intranasal drug delivery Dynamics: Extracellular and intracellular pathways revealed by Fluoro-Gold tracer in a mouse model. Brain Res 2025; 1858:149644. [PMID: 40233876 DOI: 10.1016/j.brainres.2025.149644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 04/07/2025] [Accepted: 04/11/2025] [Indexed: 04/17/2025]
Abstract
To address the challenge of drug delivery to the brain, nasal administration was introduced as a non-invasive approach to bypass the blood-brain-barrier. Intranasal (IN)-delivery routes primarily rely on the olfactory nerve and trigeminal nerve. Recent evidence of glymphatic system suggests that the perivascular space (PVS) plays a role in facilitating the rapid distribution of drugs throughout the brain. While several studies have investigated IN-delivery to the brain and have shown the presence of tracers in PVS, none have visualized PVS or trigeminal nerve in different timing or utilized Fluoro-Gold (FG) as a tracer to demonstrate associated pathways. In this study, we utilized retrograde tracer FG to illustrate the intracellular and extracellular pathways to the brain by IN-delivery and transcranial olfactory bulb (OB) injection at different time points in a mouse model. We found FG reached the outermost layer of the cerebral cortex within 30 minutes and penetrated through PVS deep into the brain over time after IN administration, possibly through extracellular pathway; similarly, FG rapidly reached the trigeminal nerve and exhibited a gradual increase in fluorescence signal at the perineural space over time. Immunofluorescence staining confirmed FG and astrocyte co-localized at the cerebral cortex and along the PVS entering the brain parenchyma. The intracellular delivery after transcranial OB injection also revealed a slow velocity of FG projection. We concluded rapid IN delivery of FG relied on the extracellular pathway, penetrating deep into the brain through glymphatic system over time. FG proved to be an excellent tool for evaluating the IN-delivery animal model, providing insights into extracellular transport via olfactory and trigeminal nerves.
Collapse
Affiliation(s)
- Meng-Ting Lin
- Department of Biomedical Engineering, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tsai-Yun Chan
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wei-Hao Liao
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chueh-Hung Wu
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan; Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| | - Tai-Horng Young
- Department of Biomedical Engineering, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Wen-Shiang Chen
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
2
|
Almahmoud A, Parekh HS, Paterson BM, Tupally KR, Vegh V. Intranasal delivery of imaging agents to the brain. Theranostics 2024; 14:5022-5101. [PMID: 39267777 PMCID: PMC11388076 DOI: 10.7150/thno.98473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 08/08/2024] [Indexed: 09/15/2024] Open
Abstract
The potential of intranasal administered imaging agents to altogether bypass the blood-brain barrier offers a promising non-invasive approach for delivery directly to the brain. This review provides a comprehensive analysis of the advancements and challenges of delivering neuroimaging agents to the brain by way of the intranasal route, focusing on the various imaging modalities and their applications in central nervous system diagnostics and therapeutics. The various imaging modalities provide distinct insights into the pharmacokinetics, biodistribution, and specific interactions of imaging agents within the brain, facilitated by the use of tailored tracers and contrast agents. Methods: A comprehensive literature search spanned PubMed, Scopus, Embase, and Web of Science, covering publications from 1989 to 2024 inclusive. Starting with advancements in tracer development, we going to explore the rationale for integration of imaging techniques, and the critical role novel formulations such as nanoparticles, nano- and micro-emulsions in enhancing imaging agent delivery and visualisation. Results: The review highlights the use of innovative formulations in improving intranasal administration of neuroimaging agents, showcasing their ability to navigate the complex anatomical and physiological barriers of the nose-to-brain pathway. Various imaging techniques, MRI, PET, SPECT, CT, FUS and OI, were evaluated for their effectiveness in tracking these agents. The findings indicate significant improvements in brain targeting efficiency, rapid uptake, and sustained brain presence using innovative formulations. Conclusion: Future directions involve the development of optimised tracers tailored for intranasal administration, the potential of multimodal imaging approaches, and the implications of these advancements for diagnosing and treating neurological disorders.
Collapse
Affiliation(s)
- Abdallah Almahmoud
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- Department of Allied Medical Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Harendra S Parekh
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
| | - Brett M Paterson
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
| | | | - Viktor Vegh
- Centre for Advanced Imaging, Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, Australia
- ARC Training Centre for Innovation in Biomedical Imaging Technology, Brisbane, QLD, Australia
| |
Collapse
|
3
|
Kaps J, Georgieva VS, Oberholz L, Kribs A, Brachvogel B, Keller T. Human preterm colostrum stimulates outgrowth in neurogenic tissue. Pediatr Res 2023; 94:1906-1910. [PMID: 37433903 PMCID: PMC10665184 DOI: 10.1038/s41390-023-02721-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 05/16/2023] [Accepted: 06/15/2023] [Indexed: 07/13/2023]
Abstract
BACKGROUND The olfactory bulb has a key role for nasal delivery of drugs to the brain by its access from the nasal mucosa and its connection to the subventricular zone. The aim of this study was to investigate the neuromodulatory capacity of human milk of premature infants on the olfactory bulb. METHODS Olfactory bulbs from P1 mice were embedded in a collagen I gel and incubated with DMEM supplemented with the aqueous phase of human colostrum (Col) of five mothers after very preterm birth, mature milk (Mat) of the same mothers or without supplement (Ctrl). After 7 days, the neurite outgrowth was quantified. Proteome analysis of the milk samples was performed using unlabeled mass spectrometry. RESULTS Outgrowth increased significantly in bulbs exposed to Col but not when exposed to Mat. Mass spectrometry revealed profound differences in the proteome of Col versus Mat. Among 21 upregulated proteins in Col were proteins involved in neurite outgrowth, axon guidance, neuromodulation and longevity. CONCLUSIONS A high bioactivity of human preterm colostrum on murine neonatal neurogenic tissue is demonstrated to be associated with a proteome profoundly differing from mature milk. IMPACT The hypothesis has been raised that neonatal brain damage in a preterm infant could potentially be ameliorated by intranasal application of maternal breast milk. In an in-vitro model using neonatal murine olfactory bulb explants a significant stimulatory effect by human preterm colostrum is observed. Proteomics reveals upregulated neuroactive proteins in human colostrum compared to mature milk. A confirmation of this exploratory study would indicate that preterm colostrum stimulates neurogenic tissue. Early intranasal colostrum application might attenuate perinatal loss of neurogenic tissue thereby contributing to reducing complications such as cerebral palsy.
Collapse
Affiliation(s)
- Julian Kaps
- Department of Pediatrics and Adolescent Medicine, Medical Faculty of the University of Cologne, Experimental Neonatology, Cologne, Germany
| | - Veronica S Georgieva
- Department of Pediatrics and Adolescent Medicine, Medical Faculty of the University of Cologne, Experimental Neonatology, Cologne, Germany
- Center for Biochemistry, Medical Faculty of the University of Cologne, Cologne, Germany
| | - Laura Oberholz
- Department of Pediatrics and Adolescent Medicine, Center for Neonatology and Pediatric Intensive Care Medicine, Medical Faculty of the University of Cologne, Cologne, Germany
| | - Angela Kribs
- Department of Pediatrics and Adolescent Medicine, Center for Neonatology and Pediatric Intensive Care Medicine, Medical Faculty of the University of Cologne, Cologne, Germany
| | - Bent Brachvogel
- Department of Pediatrics and Adolescent Medicine, Medical Faculty of the University of Cologne, Experimental Neonatology, Cologne, Germany
- Center for Biochemistry, Medical Faculty of the University of Cologne, Cologne, Germany
| | - Titus Keller
- Department of Pediatrics and Adolescent Medicine, Medical Faculty of the University of Cologne, Experimental Neonatology, Cologne, Germany.
- Department of Pediatrics and Adolescent Medicine, Center for Neonatology and Pediatric Intensive Care Medicine, Medical Faculty of the University of Cologne, Cologne, Germany.
| |
Collapse
|
4
|
Rohn TT, Radin D, Brandmeyer T, Linder BJ, Andriambeloson E, Wagner S, Kehler J, Vasileva A, Wang H, Mee JL, Fallon JH. Genetic modulation of the HTR2A gene reduces anxiety-related behavior in mice. PNAS NEXUS 2023; 2:pgad170. [PMID: 37346271 PMCID: PMC10281383 DOI: 10.1093/pnasnexus/pgad170] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 05/15/2023] [Indexed: 06/23/2023]
Abstract
The expanding field of precision gene editing using CRISPR/Cas9 has demonstrated its potential as a transformative technology in the treatment of various diseases. However, whether this genome-editing tool could be used to modify neural circuits in the central nervous system (CNS), which are implicated in complex behavioral traits, remains uncertain. In this study, we demonstrate the feasibility of noninvasive, intranasal delivery of adeno-associated virus serotype 9 (AAV9) vectors containing CRISPR/Cas9 cargo within the CNS resulting in modification of the HTR2A receptor gene. In vitro, exposure to primary mouse cortical neurons to AAV9 vectors targeting the HT2RA gene led to a concentration-dependent decrease in spontaneous electrical activity following multielectrode array (MEA) analysis. In vivo, at 5 weeks postintranasal delivery in mice, analysis of brain samples revealed single base pair deletions and nonsense mutations, leading to an 8.46-fold reduction in mRNA expression and a corresponding 68% decrease in the 5HT-2A receptor staining. Our findings also demonstrate a significant decrease in anxiety-like behavior in treated mice. This study constitutes the first successful demonstration of a noninvasive CRISPR/Cas9 delivery platform, capable of bypassing the blood-brain barrier and enabling modulation of neuronal 5HT-2A receptor pathways. The results of this study targeting the HTR2A gene provide a foundation for the development of innovative therapeutic strategies for a broad range of neurological disorders, including anxiety, depression, attentional deficits, and cognitive dysfunction.
Collapse
|
5
|
Labusek N, Mouloud Y, Köster C, Diesterbeck E, Tertel T, Wiek C, Hanenberg H, Horn PA, Felderhoff-Müser U, Bendix I, Giebel B, Herz J. Extracellular vesicles from immortalized mesenchymal stromal cells protect against neonatal hypoxic-ischemic brain injury. Inflamm Regen 2023; 43:24. [PMID: 37069694 PMCID: PMC10108458 DOI: 10.1186/s41232-023-00274-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/26/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND Human mesenchymal stromal cell (MSC)-derived extracellular vesicles (EV) revealed neuroprotective potentials in various brain injury models, including neonatal encephalopathy caused by hypoxia-ischemia (HI). However, for clinical translation of an MSC-EV therapy, scaled manufacturing strategies are required, which is challenging with primary MSCs due to inter- and intra-donor heterogeneities. Therefore, we established a clonally expanded and immortalized human MSC line (ciMSC) and compared the neuroprotective potential of their EVs with EVs from primary MSCs in a murine model of HI-induced brain injury. In vivo activities of ciMSC-EVs were comprehensively characterized according to their proposed multimodal mechanisms of action. METHODS Nine-day-old C57BL/6 mice were exposed to HI followed by repetitive intranasal delivery of primary MSC-EVs or ciMSC-EVs 1, 3, and 5 days after HI. Sham-operated animals served as healthy controls. To compare neuroprotective effects of both EV preparations, total and regional brain atrophy was assessed by cresyl-violet-staining 7 days after HI. Immunohistochemistry, western blot, and real-time PCR were performed to investigate neuroinflammatory and regenerative processes. The amount of peripheral inflammatory mediators was evaluated by multiplex analyses in serum samples. RESULTS Intranasal delivery of ciMSC-EVs and primary MSC-EVs comparably protected neonatal mice from HI-induced brain tissue atrophy. Mechanistically, ciMSC-EV application reduced microglia activation and astrogliosis, endothelial activation, and leukocyte infiltration. These effects were associated with a downregulation of the pro-inflammatory cytokine IL-1 beta and an elevated expression of the anti-inflammatory cytokines IL-4 and TGF-beta in the brain, while concentrations of cytokines in the peripheral blood were not affected. ciMSC-EV-mediated anti-inflammatory effects in the brain were accompanied by an increased neural progenitor and endothelial cell proliferation, oligodendrocyte maturation, and neurotrophic growth factor expression. CONCLUSION Our data demonstrate that ciMSC-EVs conserve neuroprotective effects of primary MSC-EVs via inhibition of neuroinflammation and promotion of neuroregeneration. Since ciMSCs can overcome challenges associated with MSC heterogeneity, they appear as an ideal cell source for the scaled manufacturing of EV-based therapeutics to treat neonatal and possibly also adult brain injury.
Collapse
Affiliation(s)
- Nicole Labusek
- Department of Pediatrics I, Neonatology & Experimental Perinatal Neurosciences, Centre for Translational and Behavioral Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Yanis Mouloud
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Christian Köster
- Department of Pediatrics I, Neonatology & Experimental Perinatal Neurosciences, Centre for Translational and Behavioral Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Eva Diesterbeck
- Department of Pediatrics I, Neonatology & Experimental Perinatal Neurosciences, Centre for Translational and Behavioral Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Tobias Tertel
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Constanze Wiek
- Department of Otorhinolaryngology and Head/Neck Surgery, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Helmut Hanenberg
- Department of Otorhinolaryngology and Head/Neck Surgery, University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
- Department of Pediatrics III, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Peter A Horn
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ursula Felderhoff-Müser
- Department of Pediatrics I, Neonatology & Experimental Perinatal Neurosciences, Centre for Translational and Behavioral Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Ivo Bendix
- Department of Pediatrics I, Neonatology & Experimental Perinatal Neurosciences, Centre for Translational and Behavioral Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| | - Josephine Herz
- Department of Pediatrics I, Neonatology & Experimental Perinatal Neurosciences, Centre for Translational and Behavioral Sciences (C-TNBS), University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| |
Collapse
|
6
|
Marcello E, Chiono V. Biomaterials-Enhanced Intranasal Delivery of Drugs as a Direct Route for Brain Targeting. Int J Mol Sci 2023; 24:ijms24043390. [PMID: 36834804 PMCID: PMC9964911 DOI: 10.3390/ijms24043390] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/22/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023] Open
Abstract
Intranasal (IN) drug delivery is a non-invasive and effective route for the administration of drugs to the brain at pharmacologically relevant concentrations, bypassing the blood-brain barrier (BBB) and minimizing adverse side effects. IN drug delivery can be particularly promising for the treatment of neurodegenerative diseases. The drug delivery mechanism involves the initial drug penetration through the nasal epithelial barrier, followed by drug diffusion in the perivascular or perineural spaces along the olfactory or trigeminal nerves, and final extracellular diffusion throughout the brain. A part of the drug may be lost by drainage through the lymphatic system, while a part may even enter the systemic circulation and reach the brain by crossing the BBB. Alternatively, drugs can be directly transported to the brain by axons of the olfactory nerve. To improve the effectiveness of drug delivery to the brain by the IN route, various types of nanocarriers and hydrogels and their combinations have been proposed. This review paper analyzes the main biomaterials-based strategies to enhance IN drug delivery to the brain, outlining unsolved challenges and proposing ways to address them.
Collapse
Affiliation(s)
- Elena Marcello
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- Interuniversity Center for the Promotion of 3Rs Principles in Teaching and Research, Centro 3R, 56122 Pisa, Italy
| | - Valeria Chiono
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Corso Duca Degli Abruzzi 24, 10129 Turin, Italy
- Interuniversity Center for the Promotion of 3Rs Principles in Teaching and Research, Centro 3R, 56122 Pisa, Italy
- Institute for Chemical-Physical Processes, National Research Council (CNR-IPCF), 56124 Pisa, Italy
- Correspondence:
| |
Collapse
|
7
|
Crowe TP, Hsu WH. Evaluation of Recent Intranasal Drug Delivery Systems to the Central Nervous System. Pharmaceutics 2022; 14:629. [PMID: 35336004 PMCID: PMC8950509 DOI: 10.3390/pharmaceutics14030629] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 01/27/2023] Open
Abstract
Neurological diseases continue to increase in prevalence worldwide. Combined with the lack of modifiable risk factors or strongly efficacious therapies, these disorders pose a significant and growing burden on healthcare systems and societies. The development of neuroprotective or curative therapies is limited by a variety of factors, but none more than the highly selective blood-brain barrier. Intranasal administration can bypass this barrier completely and allow direct access to brain tissues, enabling a large number of potential new therapies ranging from bioactive peptides to stem cells. Current research indicates that merely administering simple solutions is inefficient and may limit therapeutic success. While many therapies can be delivered to some degree without carrier molecules or significant modification, a growing body of research has indicated several methods of improving the safety and efficacy of this administration route, such as nasal permeability enhancers, gelling agents, or nanocarrier formulations. This review shall discuss promising delivery systems and their role in expanding the clinical efficacy of this novel administration route. Optimization of intranasal administration will be crucial as novel therapies continue to be studied in clinical trials and approved to meet the growing demand for the treatment of patients with neurological diseases.
Collapse
Affiliation(s)
- Tyler P. Crowe
- Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA;
| | - Walter H. Hsu
- Department of Biomedical Sciences, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
8
|
Abstract
RNA-based therapeutics have entered the mainstream with seemingly limitless possibilities to treat all categories of neurological disease. Here, common RNA-based drug modalities such as antisense oligonucleotides, small interfering RNAs, RNA aptamers, RNA-based vaccines and mRNA drugs are reviewed highlighting their current and potential applications. Rapid progress has been made across rare genetic diseases and neurodegenerative disorders, but safe and effective delivery to the brain remains a significant challenge for many applications. The advent of individualized RNA-based therapies for ultra-rare diseases is discussed against the backdrop of the emergence of this field into more common conditions such as Alzheimer's disease and ischaemic stroke. There remains significant untapped potential in the use of RNA-based therapeutics for behavioural disorders and tumours of the central nervous system; coupled with the accelerated development expected over the next decade, the true potential of RNA-based therapeutics to transform the therapeutic landscape in neurology remains to be uncovered.
Collapse
Affiliation(s)
- Karen Anthony
- Centre for Physical Activity and Life Sciences, University of Northampton, Northampton, UK
| |
Collapse
|
9
|
Bahlakeh G, Rahbarghazi R, Mohammadnejad D, Abedelahi A, Karimipour M. Current knowledge and challenges associated with targeted delivery of neurotrophic factors into the central nervous system: focus on available approaches. Cell Biosci 2021; 11:181. [PMID: 34641969 PMCID: PMC8507154 DOI: 10.1186/s13578-021-00694-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/28/2021] [Indexed: 12/23/2022] Open
Abstract
During the last decades, numerous basic and clinical studies have been conducted to assess the delivery efficiency of therapeutic agents into the brain and spinal cord parenchyma using several administration routes. Among conventional and in-progress administrative routes, the eligibility of stem cells, viral vectors, and biomaterial systems have been shown in the delivery of NTFs. Despite these manifold advances, the close association between the delivery system and regeneration outcome remains unclear. Herein, we aimed to discuss recent progress in the delivery of these factors and the pros and cons related to each modality.
Collapse
Affiliation(s)
- Gozal Bahlakeh
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Daruosh Mohammadnejad
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Abedelahi
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Karimipour
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran. .,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
10
|
Palmieri B, Corazzari V, Vadalaʹ M, Vallelunga A, Morales-Medina JC, Iannitti T. The role of sensory and olfactory pathways in multiple chemical sensitivity. REVIEWS ON ENVIRONMENTAL HEALTH 2021; 36:319-326. [PMID: 33070122 DOI: 10.1515/reveh-2020-0058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 09/15/2020] [Indexed: 06/11/2023]
Abstract
Multiple chemical sensitivity (MCS) is characterised by non-specific and recurring symptoms affecting multiple organs and associated with exposure to chemicals, even at low concentrations, which are, under normal circumstances, harmless to the general population. Symptoms include general discomfort, cardiovascular instability, irritation of the sensory organs, breath disorders, hypersensitivity affecting the skin and epithelial lining of the gut, throat and lungs, anxiety, and learning and memory loss. Chemical intolerance is a key distinguishing feature of MCS, limiting considerably patients' lifestyle with serious social, occupational and economic implications. Since no specific diagnostic markers are currently available for chemical intolerance, the diagnosis relies on clinical symptoms. Despite the formulation of several hypotheses regarding the pathophysiology of MCS, its mechanisms remain undefined. A person-centred care approach, based on multidisciplinary and individualised medical plans, has shown promising results. However, more definite treatment strategies are required. We have reviewed the main experimental studies on MCS pathophysiology, focusing on the brain networks involved, the impact of environmental pollution on the olfactory system and the correlation with other pathologies such as neurodegenerative diseases. Finally, we discuss treatment strategies targeting the olfactory system.
Collapse
Affiliation(s)
- Beniamino Palmieri
- Surgical, Medical, Dental and Morphological Sciences Departments with interest in Transplants, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Network of Second Opinion, Modena, Italy
| | | | - Maria Vadalaʹ
- Surgical, Medical, Dental and Morphological Sciences Departments with interest in Transplants, Oncology and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
- Network of Second Opinion, Modena, Italy
| | - Annamaria Vallelunga
- Department of Medicine and Surgery, Centre for Neurodegenerative Diseases (CEMAND), University of Salerno, Salerno, Italy
| | - Julio César Morales-Medina
- Centro de Investigación en Reproducción Animal, CINVESTAV- Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | - Tommaso Iannitti
- Charles River Discovery Research Services UK Limited, Portishead, UK
| |
Collapse
|
11
|
Erichsen JM, Calva CB, Reagan LP, Fadel JR. Intranasal insulin and orexins to treat age-related cognitive decline. Physiol Behav 2021; 234:113370. [PMID: 33621561 PMCID: PMC8053680 DOI: 10.1016/j.physbeh.2021.113370] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 02/19/2021] [Indexed: 02/06/2023]
Abstract
The intranasal (IN) administration of neuropeptides, such as insulin and orexins, has been suggested as a treatment strategy for age-related cognitive decline (ARCD). Because dysfunctional neuropeptide signaling is an observed characteristic of ARCD, it has been suggested that IN delivery of insulin and/or orexins may restore endogenous peptide signaling and thereby preserve cognition. IN administration is particularly alluring as it is a relatively non-invasive method that directly targets peptides to the brain. Several laboratories have examined the behavioral effects of IN insulin in young, aged, and cognitively impaired rodents and humans. These studies demonstrated improved performance on various cognitive tasks following IN insulin administration. Fewer laboratories have assessed the effects of IN orexins; however, this peptide also holds promise as an effective treatment for ARCD through the activation of the cholinergic system and/or the reduction of neuroinflammation. Here, we provide a brief overview of the advantages of IN administration and the delivery pathway, then summarize the current literature on IN insulin and orexins. Additional preclinical studies will be useful to ultimately uncover the mechanisms underlying the pro-cognitive effects of IN insulin and orexins, whereas future clinical studies will aid in the determination of the most efficacious dose and dosing paradigm. Eventually, IN insulin and/or orexin administration may be a widely used treatment strategy in the clinic for ARCD.
Collapse
Affiliation(s)
- Jennifer M Erichsen
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC 29208, United States.
| | - Coleman B Calva
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC 29208, United States
| | - Lawrence P Reagan
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC 29208, United States; Columbia VA Health Care System, Columbia, SC, 29208, United States
| | - Jim R Fadel
- University of South Carolina School of Medicine, Department of Pharmacology, Physiology, and Neuroscience, Columbia, SC 29208, United States
| |
Collapse
|
12
|
Lebedev AA, Bessolova YN, Efimov NS, Bychkov ER, Droblenkov AV, Shabanov PD. Role of orexin peptide system in emotional overeating induced by brain reward stimulation in fed rats. RESEARCH RESULTS IN PHARMACOLOGY 2020. [DOI: 10.3897/rrpharmacology.6.52180] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Introduction: The purpose of this work was to prove that the reaction of food self-deprivation in “fed up” rats is a suitable model for studying the emotional overeating in the experiment.
Methods: The self-deprivation reaction, i.e. self-isolation of an animal from food during electrical self-stimulation of the brain, was studied in animals with food deprivation. To reproduce the self-stimulation of the lateral hypothalamus, the male Wistar rats were trained to press a pedal in a Skinner box. After training, the rats received food deprivation, then a feeder was placed in the Skinner box, and a conditioned food reflex was developed in rats within 5 days.
Results and discussion: The food self-deprivation reaction was observed in the ”satiated” rats with a current intensity of 10% and above the threshold for self-stimulation. Hungry animals pressed the pedal for hypothalamic self-stimulation and took no notice of the feeding trough. Sulpiride, a dopamine D2 antagonist (5 and 20 mg/kg i.p.), administered to the “satiated” rats decreased both the eating behavior and self-stimulation in food self-deprivation testing. SB-408124, an orexin A receptor antagonist (0.5 mg/ml, 20 μl intranasally) reduced only the number of pellets eaten, but not the number of pedal presses.
Conclusion: The orexin A receptors are preferably involved in emotional eating compared with orexin B (OX2R TCS-OX2-29) and D2 dopamine receptors. Because emotional eating is significantly related to clinical eating disorders, like bulimia and binge eating disorder, it seems promising to use drugs of the orexin system to treat and prevent the issue.
Collapse
|
13
|
Casadomé-Perales Á, Matteis LD, Alleva M, Infantes-Rodríguez C, Palomares-Pérez I, Saito T, Saido TC, Esteban JA, Nebreda AR, de la Fuente JM, Dotti CG. Inhibition of p38 MAPK in the brain through nasal administration of p38 inhibitor loaded in chitosan nanocapsules. Nanomedicine (Lond) 2019; 14:2409-2422. [PMID: 31456488 DOI: 10.2217/nnm-2018-0496] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Aim: To determine whether a p38 MAPK inhibitor incorporated into nanoemulsion-based chitosan nanocapsules can reduce the activity of this kinase in the brain through their nasal administration in mice. Materials & methods: We selected the p38 MAPK inhibitor PH797804, an ATP-competitive inhibitor of p38α encapsulated in nanoemulsion-based chitosan nanocapsules. Biological effect was evaluated in microglial and neuronal cells in vitro and in ex vivo and in vivo systems, in a mouse model of Alzheimer's disease. Results: Encapsulated inhibitor retains enzymatic inhibitory activity and tissue penetration capacity in vitro, ex vivo and in vivo. Conclusion: Nasal administration of chitosan nanocapsules can be an effective approach for brain-restricted reduction of p38 MAPK activity, thus reducing the side effects of systemic administration.
Collapse
Affiliation(s)
- Álvaro Casadomé-Perales
- Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa, CSIC/UAM, 28049 Madrid, Spain
| | - Laura De Matteis
- Instituto de Nanociencia de Aragón (INA), Universidad de Zaragoza & CIBER-BBN, 50018 Zaragoza, Spain
| | - Maria Alleva
- Instituto de Nanociencia de Aragón (INA), Universidad de Zaragoza & CIBER-BBN, 50018 Zaragoza, Spain
| | - Cristina Infantes-Rodríguez
- Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa, CSIC/UAM, 28049 Madrid, Spain
| | - Irene Palomares-Pérez
- Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa, CSIC/UAM, 28049 Madrid, Spain
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama 351-0106, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama 351-0106, Japan
| | - José A Esteban
- Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa, CSIC/UAM, 28049 Madrid, Spain
| | - Angel R Nebreda
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science & Technology, & ICREA, 08028 Barcelona, Spain
| | - Jesús M de la Fuente
- Instituto de Ciencia de Materiales de Aragón (ICMA), CSIC-Universidad de Zaragoza & CIBER-BBN, 50018 Zaragoza, Spain
| | - Carlos G Dotti
- Department of Molecular Neuropathology, Centro de Biología Molecular Severo Ochoa, CSIC/UAM, 28049 Madrid, Spain
| |
Collapse
|
14
|
Nervous system delivery of antilysophosphatidic acid antibody by nasal application attenuates mechanical allodynia after traumatic brain injury in rats. Pain 2018; 158:2181-2188. [PMID: 29028747 DOI: 10.1097/j.pain.0000000000001019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Lysophosphatidic acid (LPA) is a bioactive lipid that impacts neurological outcomes after neurotrauma by inhibiting neuroregeneration, promoting inflammation, and contributing to behavioral deficits. Blocking LPA signaling with a novel anti-LPA monoclonal antibody (mAb) is neuroprotective after traumatic brain injury (TBI) if given to injured animals whose blood-brain barrier (BBB) has been compromised. It is hypothesized that the anti-LPA mAb could improve chronic pain initiated by TBI. However, poor brain penetration after systemic application of the antibody makes access to the central nervous system (CNS) problematic in situations where the BBB is intact. Our experiments investigated whether intranasal delivery of the anti-LPA mAb could bypass the BBB, allowing for direct entry of the antibody to certain areas of the CNS. When the humanized anti-LPA mAb, LT3114, was intranasally applied to injured rats within 30 minutes after mild TBI using the central lateral percussion model, enzyme-linked immunospecific assay and immunohistochemistry demonstrated antibody uptake to several areas in the CNS, including the area of cortical injury, the corpus callosum, cerebellum, and the subventricular region. Compared with control rats that received LT3114 but no TBI, TBI rats demonstrated significantly higher concentrations of intranasally administered LT3114 antibody in some tissues. In behavioral studies, a significant attenuation of mechanical allodynia after TBI was observed in the anti-LPA treatment group (P = 0.0079), when compared with vehicle controls within 14 days after TBI. These results suggest that intranasal application of the anti-LPA antibody directly accesses CNS sites involved in TBI-related pain and that this access attenuates pain sequelae to the neurotrauma.
Collapse
|
15
|
Chen N, Chopp M, Xiong Y, Qian JY, Lu M, Zhou D, He L, Liu Z. Subacute intranasal administration of tissue plasminogen activator improves stroke recovery by inducing axonal remodeling in mice. Exp Neurol 2018. [PMID: 29518364 DOI: 10.1016/j.expneurol.2018.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In addition to thrombolysis, tissue plasminogen activator (tPA) can evoke neurorestorative processes. We therefore investigated the therapeutic effect of subacute intranasal administration of tPA post stroke on neurological recovery and on corticospinal innervation in mice. A transgenic mouse line, in which the pyramidal neurons and corticospinal tract (CST) axons are specifically labeled by yellow fluorescent protein (YFP) was employed. Adult CST-YFP mice were subjected to right unilateral middle cerebral artery occlusion (MCAo), and were randomly divided into groups treated with saline or tPA intranasally in the subacute phase. Pseudorabies virus (PRV)-614-monomeric red fluorescent protein (RFP) was injected into the left forelimb. The cervical spinal cord and brain were processed for fluorescent microscopy to detect YFP and RFP labeling. Primary embryonic neurons were cultured with tPA at different concentrations. Neurite length and branch numbers were then measured. In vivo, subacute tPA treatment significantly enhanced functional recovery (p < 0.05), and increased CST density in the denervated gray matter, and in the numbers of PRV-labeled neurons in bilateral cortices. The behavioral performance was significantly correlated with axonal density in the denervated spinal cord. In vitro, both neurite length and branch numbers significantly increased with concentration of tPA (p < 0.05). Our results demonstrate that tPA dose-dependently increases neurite outgrowth and branching of cultured cortical neurons. Subacute intranasal administration of tPA may provide enhance neurological recovery after stroke by promoting CST axonal remodeling.
Collapse
Affiliation(s)
- Ning Chen
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, PR China; Department of Neurology, Henry Ford Hospital, Detrot, MI, United States
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detrot, MI, United States; Department of Physics, Oakland University, Rochester, MI, United States
| | - Ye Xiong
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI, United States
| | - Jian-Yong Qian
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, PR China
| | - Mei Lu
- Biostatistics and Research Epidemiology, Henry Ford Hospital, Detroit, MI, United States
| | - Dong Zhou
- Department of Neurology, Henry Ford Hospital, Detrot, MI, United States
| | - Li He
- Department of Neurology, Henry Ford Hospital, Detrot, MI, United States.
| | - Zhongwu Liu
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, Sichuan, PR China.
| |
Collapse
|
16
|
Mechanism of intranasal drug delivery directly to the brain. Life Sci 2018; 195:44-52. [DOI: 10.1016/j.lfs.2017.12.025] [Citation(s) in RCA: 266] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 12/08/2017] [Accepted: 12/19/2017] [Indexed: 01/09/2023]
|
17
|
Glufosinate aerogenic exposure induces glutamate and IL-1 receptor dependent lung inflammation. Clin Sci (Lond) 2016; 130:1939-54. [PMID: 27549113 DOI: 10.1042/cs20160530] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 08/22/2016] [Indexed: 11/17/2022]
Abstract
Glufosinate-ammonium (GLA), the active component of an herbicide, is known to cause neurotoxicity. GLA shares structural analogy with glutamate. It is a powerful inhibitor of glutamine synthetase (GS) and may bind to glutamate receptors. Since these potentials targets of GLA are present in lung and immune cells, we asked whether airway exposure to GLA may cause lung inflammation in mice. A single GLA exposure (1 mg/kg) induced seizures and inflammatory cell recruitment in the broncho-alveolar space, and increased myeloperoxidase (MPO), inducible NO synthase (iNOS), interstitial inflammation and disruption of alveolar septae within 6-24 h. Interleukin 1β (IL-1β) was increased and lung inflammation depended on IL-1 receptor 1 (IL-1R1). We demonstrate that glutamate receptor pathway is central, since the N-methyl-D-aspartate (NMDA) receptor inhibitor MK-801 prevented GLA-induced lung inflammation. Chronic exposure (0.2 mg/kg 3× per week for 4 weeks) caused moderate lung inflammation and enhanced airway hyperreactivity with significant increased airway resistance. In conclusion, GLA aerosol exposure causes glutamate signalling and IL-1R-dependent pulmonary inflammation with airway hyperreactivity in mice.
Collapse
|
18
|
Abstract
Although an association between diabetes mellitus (DM) and cognitive dysfunction has been recognized for a century, it is often not considered as a complication of DM and remains under-recognized. Cognitive dysfunction, usually present as mild cognitive impairment, can occur with either type 1 or type 2 DM. Both forms of DM contribute to accelerated cerebral atrophy and to the presence of heightened white matter abnormalities. These effects are noted most at the two extremes of life, in childhood and in the advanced years. The cognitive spheres most affected include attention and executive function, processing speed, perception, and memory. Although DM is unlikely to lead to frank dementia, its ability to exacerbate existing neurodegenerative processes, such as Alzheimer disease, will impact tremendously upon our society in the upcoming decades as our population ages. This chapter describes the clinical impact of DM upon the brain, along with discussion of the potential therapeutic avenues to be discovered in the coming decades. We need to prepare for better preventative and therapeutic management of this cerebral neurodegenerative condition.
Collapse
Affiliation(s)
- Cory Toth
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
19
|
Endres K, Reinhardt S, Geladaris A, Knies J, Grimm M, Hartmann T, Schmitt U. Transnasal delivery of human A-beta peptides elicits impaired learning and memory performance in wild type mice. BMC Neurosci 2016; 17:44. [PMID: 27377996 PMCID: PMC4932715 DOI: 10.1186/s12868-016-0280-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 06/24/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Murine models of Alzheimer's disease (AD) are mainly based on overexpression of pathologic amyloid precursor protein and/or presenilins. Those genes resemble underlying cause of early onset type of AD while about 99 % of all human cases are to be characterized as sporadic, late onset. Appropriate animal models for this type of AD are still missing. We here investigated, if transnasal delivery of A-beta 42 peptides might serve to mimic pathological effects in mice. RESULTS A-beta 42 peptides, used for the behavioral study, showed the expected dose-dependent toxicity in neur oblastoma cell line SH-SY5Y and were able to form higher molecular weight species in vitro. Upon delivery into nostrils of wild type mice, protein bands that might represent aggregation products of the exogenously applied human A-beta 42 were only observed in total brain homogenates from mice pre-treated with mannitol. By using TAMRA-labeled A-beta 42 peptides we demonstrated, that transport throughout the brain was achieved already 1 h after administration. FVB/N mice treated with A-beta 42 for 3 days were significantly impaired in the cue-retention condition of the fear conditioning task as compared to controls whereas A-beta-treated C57B6/J mice were impaired in the context condition. In the Morris water maze test, these mice also displayed a delayed learning performance, indicated by significantly longer time to find the platform. Those deficits were also seen for memory performance in the probe trial as measured by number of crossings of the former platform position and time spent in the goal quadrant. CONCLUSIONS Existing AD mouse models are of genetic origin and need prolonged housing time before onset of pathology. Our short-term treatment induced learning and memory deficits via exogenous application of A-beta peptides comparable to those observed for the transgenic animals. With the transnasal A-beta 42 treatment we present an approach to investigate purely A-beta related changes suitable as a model for symptoms of Alzheimer's dementia (AD). Resulting behavioral deficits were indicative for familial type of Alzheimer's disease as well as for the late onset variant.
Collapse
Affiliation(s)
- Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg University, Untere Zahlbacher Straße 8, 55131, Mainz, Germany.
| | - Sven Reinhardt
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg University, Untere Zahlbacher Straße 8, 55131, Mainz, Germany
| | - Anastasia Geladaris
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg University, Untere Zahlbacher Straße 8, 55131, Mainz, Germany
| | - Julia Knies
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg University, Untere Zahlbacher Straße 8, 55131, Mainz, Germany
| | - Marcus Grimm
- Deutsches Institut für DemenzPrävention (DIDP), Neurodegeneration and Neurobiology, Saarland University, Homburg/Saar, Germany.,Experimental Neurology, Saarland University, Homburg/Saar, Germany
| | - Tobias Hartmann
- Deutsches Institut für DemenzPrävention (DIDP), Neurodegeneration and Neurobiology, Saarland University, Homburg/Saar, Germany.,Experimental Neurology, Saarland University, Homburg/Saar, Germany
| | - Ulrich Schmitt
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg University, Untere Zahlbacher Straße 8, 55131, Mainz, Germany
| |
Collapse
|
20
|
Guardia Clausi M, Paez P, Pasquini L, Pasquini J. Inhalation of growth factors and apo-transferrin to protect and repair the hypoxic-ischemic brain. Pharmacol Res 2016; 109:81-5. [DOI: 10.1016/j.phrs.2016.01.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 01/12/2016] [Accepted: 01/12/2016] [Indexed: 10/22/2022]
|
21
|
Linares GR, Chiu CT, Scheuing L, Leng Y, Liao HM, Maric D, Chuang DM. Preconditioning mesenchymal stem cells with the mood stabilizers lithium and valproic acid enhances therapeutic efficacy in a mouse model of Huntington's disease. Exp Neurol 2016; 281:81-92. [DOI: 10.1016/j.expneurol.2016.04.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 04/02/2016] [Accepted: 04/04/2016] [Indexed: 01/30/2023]
|
22
|
Kim ID, Sawicki E, Lee HK, Lee EH, Park HJ, Han PL, Kim KK, Choi H, Lee JK. Robust neuroprotective effects of intranasally delivered iNOS siRNA encapsulated in gelatin nanoparticles in the postischemic brain. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:1219-29. [PMID: 26945975 DOI: 10.1016/j.nano.2016.01.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 12/10/2015] [Accepted: 01/05/2016] [Indexed: 10/22/2022]
Abstract
The therapeutic efficacy of intranasal iNOS siRNA delivery was investigated in the postischemic rat brain after encapsulating on in gelatin nanoparticles (GNPs; diameter 188.0 ± 60.9 nm) cross-linked with 0.0667% glutaraldehyde (GA). Intranasally delivered GNPs were found in extracellular and intracellular compartments of many brain regions, including the olfactory bulb, cerebral cortex, and striatum at 1 hour after infusion and continued to be detected for days. Infarct volumes were markedly suppressed (maximal reduction to 42.1 ± 2.6%) at 2 days after 60 minutes of middle cerebral artery occlusion (MCAO) when iNOS siRNA/GNPs were delivered at 6 hours post-MCAO. In addition, this protective effect was manifested by reductions in neurological and behavioral deficits that were sustained for 2 weeks. Therapeutic potency of iNOS siRNA/GNPs was significantly greater and sustained longer than that of bare siRNA and prolonged and efficient iNOS by iNOS siRNA/GNP is responsible for the robust neuroprotective effect.
Collapse
Affiliation(s)
- Il-Doo Kim
- Department of Anatomy, Inha University School of Medicine, Inchon, Korea; Medical Research Center, Inha University School of Medicine, Inchon, Korea
| | - Elizabeth Sawicki
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Hye-Kyung Lee
- Department of Anatomy, Inha University School of Medicine, Inchon, Korea; Medical Research Center, Inha University School of Medicine, Inchon, Korea
| | - Eun-Hwa Lee
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Heon Joo Park
- Medical Research Center, Inha University School of Medicine, Inchon, Korea; Department of Microbiology, Inha University School of Medicine, Inchon, Korea
| | - Pyung-Lim Han
- Department of Brain and Cognitive Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Kyekyoon Kevin Kim
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA; Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL USA
| | - Hyungsoo Choi
- Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, IL USA
| | - Ja-Kyeong Lee
- Department of Anatomy, Inha University School of Medicine, Inchon, Korea; Medical Research Center, Inha University School of Medicine, Inchon, Korea.
| |
Collapse
|
23
|
Bender T, Migliore M, Campbell R, John Gatley S, Waszczak B. Intranasal administration of glial-derived neurotrophic factor (GDNF) rapidly and significantly increases whole-brain GDNF level in rats. Neuroscience 2015; 303:569-76. [DOI: 10.1016/j.neuroscience.2015.07.016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 06/16/2015] [Accepted: 07/06/2015] [Indexed: 12/31/2022]
|
24
|
Abstract
Developing therapies for the brain is perhaps the greatest challenge facing modern medicine today. While a great many potential therapies show promise in animal models, precious few make it to approval or are even studied in human patients. The particular challenges to the translation of neurotherapeutics to the clinic are many, but a major barrier is difficulty in delivering therapeutics into the brain. The goal of this workshop was to present ways to deliver therapeutics to the brain, including the limitations of each method, and describe ways to track their delivery, safety, and efficacy. Solving the problem of delivery will aid translation of therapeutics for patients suffering from neurodegeneration and other disorders of the brain.
Collapse
Affiliation(s)
- Patricia I Dickson
- Division of Medical Genetics, Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, 1124 W. Carson Street, HH1, Torrance, CA, 90502, USA,
| |
Collapse
|
25
|
Chauhan MB, Chauhan NB. Brain Uptake of Neurotherapeutics after Intranasal versus Intraperitoneal Delivery in Mice. JOURNAL OF NEUROLOGY AND NEUROSURGERY 2015; 2:009. [PMID: 26366437 PMCID: PMC4567259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
There is a growing global prevalence of neurodegenerative diseases such as Alzheimer's disease and dementia. Current treatment for neurodegenerative diseases is limited due to the blood brain barrier's ability to restrict the entry of therapeutics to the brain. In that context, direct delivery of drugs from nose to brain has gained emerging interest as an important alternative to oral and parenteral routes of administration. Although there are considerable reports showing promising results after intranasal drug delivery in various disease-models and investigatory human clinical trials, there are very few studies showing a detailed pharmacokinetics with regard to the uptake and retention of intranasally delivered material(s) within specific brain regions, which are critical determining factors for dosing conditions and optimal treatment regimen. This investigation compared a time-dependent brain uptake and resident time of various radiolabeled candidate neurotherapeutics after a single bolus intranasal or intraperitoneal administration in mice. Results indicate that the brain uptake of intranasally delivered therapeutic(s) is > 5 times greater than that after intraperitoneal delivery. The peak uptake and resident time of all intranasally delivered test therapeutics for all brain regions is observed to be between 30min-12h, depending upon the distance of brain region from the site of administration, followed by gradual fading of radioactive counts by 24h post intranasal administration. Current study confirms the usefulness of intranasal administration as a non- invasive and efficient means of delivering therapeutics to the brain to treat neurodegenerative diseases including Alzheimer's disease.
Collapse
Affiliation(s)
| | - Neelima B. Chauhan
- Department of pediatrics, University of Illinois at Chicago, Children’s Hospital of the University of Illinois, Chicago, IL, USA
- Neuroscience Research, R & D, Jesse Brown VA Medical Center, Chicago, IL, USA
| |
Collapse
|
26
|
Neurotrophic and neuroprotective efficacy of intranasal GDNF in a rat model of Parkinson's disease. Neuroscience 2014; 274:11-23. [PMID: 24845869 DOI: 10.1016/j.neuroscience.2014.05.019] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 05/05/2014] [Accepted: 05/10/2014] [Indexed: 01/27/2023]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) exerts neurotrophic and neuroprotective effects on substantia nigra (SN) dopamine neurons and has great therapeutic potential for Parkinson's disease (PD). Hindering this potential is the fact that GDNF cannot cross the blood-brain barrier. The aim of this study was to assess the effects of GDNF administered by the intranasal route in normal rats, and in the unilateral 6-hydroxydopamine (6-OHDA) model of PD. In the first study, rats received single intranasal doses of 50-μg GDNF in phosphate-buffered saline (PBS) or cationic liposomes, but no 6-OHDA. In the second study, rats were nasally administered 10, 50 or 150 μg of GDNF in PBS or cationic liposomes 1h before injection of 6-OHDA. All groups were sacrificed 3-4 weeks later. Both intranasal GDNF treatments induced a neurotrophic effect in the SN insofar as the number of tyrosine hydroxylase (TH)-positive neurons was significantly higher than in controls given intranasal PBS liposomes. Dopamine cell counts were also higher in the intact SN of 6-OHDA-lesioned rats compared to controls given PBS liposomes. Most importantly, intranasal GDNF provided significant neuroprotective efficacy indicated by greater TH immunostaining density in the lesioned versus intact SN of rats given single 50-μg doses of GDNF in PBS, or 150-μg doses of liposomal GDNF, compared to lesioned rats given PBS liposomes. Three 50-μg doses given at daily intervals (1 day before, 1h before, and 1 day after 6-OHDA) provided even greater protection than single 150-μg doses. Multiple doses at short intervals may therefore provide greater neuroprotection than single bolus doses. These results demonstrate both a neurotrophic effect of intranasal GDNF in the intact SN as well as neuroprotective efficacy in the unilateral 6-OHDA model, supporting pursuit of this approach as a potential treatment for PD.
Collapse
|
27
|
Mohammad MG, Tsai VW, Ruitenberg MJ, Hassanpour M, Li H, Hart PH, Breit SN, Sawchenko PE, Brown DA. Immune cell trafficking from the brain maintains CNS immune tolerance. J Clin Invest 2014; 124:1228-41. [PMID: 24569378 PMCID: PMC3934177 DOI: 10.1172/jci71544] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 11/21/2013] [Indexed: 01/12/2023] Open
Abstract
In the CNS, no pathway dedicated to immune surveillance has been characterized for preventing the anti-CNS immune responses that develop in autoimmune neuroinflammatory disease. Here, we identified a pathway for immune cells to traffic from the brain that is associated with the rostral migratory stream (RMS), which is a forebrain source of newly generated neurons. Evaluation of fluorescently labeled leukocyte migration in mice revealed that DCs travel via the RMS from the CNS to the cervical LNs (CxLNs), where they present antigen to T cells. Pharmacologic interruption of immune cell traffic with the mononuclear cell-sequestering drug fingolimod influenced anti-CNS T cell responses in the CxLNs and modulated experimental autoimmune encephalomyelitis (EAE) severity in a mouse model of multiple sclerosis (MS). Fingolimod treatment also induced EAE in a disease-resistant transgenic mouse strain by altering DC-mediated Treg functions in CxLNs and disrupting CNS immune tolerance. These data describe an immune cell pathway that originates in the CNS and is capable of dampening anti-CNS immune responses in the periphery. Furthermore, these data provide insight into how fingolimod treatment might exacerbate CNS neuroinflammation in some cases and suggest that focal therapeutic interventions, outside the CNS have the potential to selectively modify anti-CNS immunity.
Collapse
MESH Headings
- Animals
- CD11 Antigens/metabolism
- Cell Movement
- Cells, Cultured
- Dendritic Cells/physiology
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Fingolimod Hydrochloride
- Immune Tolerance
- Lymph Nodes/immunology
- Lymph Nodes/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Neck
- Propylene Glycols
- Prosencephalon/immunology
- Prosencephalon/pathology
- Sphingosine/analogs & derivatives
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Mohammad G. Mohammad
- Laboratory of Neuroinflammation, St. Vincent’s Centre for Applied Medical Research and University of New South Wales, Sydney, New South Wales, Australia.
School of Biomedical Sciences and Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia.
Telethon Institute for Child Health Research, University of Western Australia, Perth, Western Australia, Australia.
Laboratory of Neuronal Structure and Function, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Vicky W.W. Tsai
- Laboratory of Neuroinflammation, St. Vincent’s Centre for Applied Medical Research and University of New South Wales, Sydney, New South Wales, Australia.
School of Biomedical Sciences and Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia.
Telethon Institute for Child Health Research, University of Western Australia, Perth, Western Australia, Australia.
Laboratory of Neuronal Structure and Function, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Marc J. Ruitenberg
- Laboratory of Neuroinflammation, St. Vincent’s Centre for Applied Medical Research and University of New South Wales, Sydney, New South Wales, Australia.
School of Biomedical Sciences and Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia.
Telethon Institute for Child Health Research, University of Western Australia, Perth, Western Australia, Australia.
Laboratory of Neuronal Structure and Function, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Masoud Hassanpour
- Laboratory of Neuroinflammation, St. Vincent’s Centre for Applied Medical Research and University of New South Wales, Sydney, New South Wales, Australia.
School of Biomedical Sciences and Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia.
Telethon Institute for Child Health Research, University of Western Australia, Perth, Western Australia, Australia.
Laboratory of Neuronal Structure and Function, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Hui Li
- Laboratory of Neuroinflammation, St. Vincent’s Centre for Applied Medical Research and University of New South Wales, Sydney, New South Wales, Australia.
School of Biomedical Sciences and Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia.
Telethon Institute for Child Health Research, University of Western Australia, Perth, Western Australia, Australia.
Laboratory of Neuronal Structure and Function, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Prue H. Hart
- Laboratory of Neuroinflammation, St. Vincent’s Centre for Applied Medical Research and University of New South Wales, Sydney, New South Wales, Australia.
School of Biomedical Sciences and Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia.
Telethon Institute for Child Health Research, University of Western Australia, Perth, Western Australia, Australia.
Laboratory of Neuronal Structure and Function, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Samuel N. Breit
- Laboratory of Neuroinflammation, St. Vincent’s Centre for Applied Medical Research and University of New South Wales, Sydney, New South Wales, Australia.
School of Biomedical Sciences and Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia.
Telethon Institute for Child Health Research, University of Western Australia, Perth, Western Australia, Australia.
Laboratory of Neuronal Structure and Function, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Paul E. Sawchenko
- Laboratory of Neuroinflammation, St. Vincent’s Centre for Applied Medical Research and University of New South Wales, Sydney, New South Wales, Australia.
School of Biomedical Sciences and Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia.
Telethon Institute for Child Health Research, University of Western Australia, Perth, Western Australia, Australia.
Laboratory of Neuronal Structure and Function, Salk Institute for Biological Studies, La Jolla, California, USA
| | - David A. Brown
- Laboratory of Neuroinflammation, St. Vincent’s Centre for Applied Medical Research and University of New South Wales, Sydney, New South Wales, Australia.
School of Biomedical Sciences and Queensland Brain Institute, University of Queensland, Brisbane, Queensland, Australia.
Telethon Institute for Child Health Research, University of Western Australia, Perth, Western Australia, Australia.
Laboratory of Neuronal Structure and Function, Salk Institute for Biological Studies, La Jolla, California, USA
| |
Collapse
|
28
|
MALDI Mass Spectrometry Imaging of 1-Methyl-4-phenylpyridinium (MPP+) in Mouse Brain. Neurotox Res 2013; 25:135-45. [DOI: 10.1007/s12640-013-9449-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Revised: 11/24/2013] [Accepted: 12/05/2013] [Indexed: 12/01/2022]
|
29
|
Xiao C, Davis FJ, Chauhan BC, Viola KL, Lacor PN, Velasco PT, Klein WL, Chauhan NB. Brain transit and ameliorative effects of intranasally delivered anti-amyloid-β oligomer antibody in 5XFAD mice. J Alzheimers Dis 2013; 35:777-88. [PMID: 23542865 DOI: 10.3233/jad-122419] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease (AD) is a global health crisis with limited treatment options. Despite major advances in neurotherapeutics, poor brain penetration due to the blood-brain barrier continues to pose a big challenge in overcoming the access of therapeutics to the central nervous system. In that regard, the non-invasive intranasal route of brain targeting is gaining considerable attention. The nasal mucosa offers a large surface area, rapid absorption, and avoidance of first-pass metabolism increasing drug bioavailability with less systemic side effects. Intranasal delivery is known to utilize olfactory, rostral migratory stream, and trigeminal routes to reach the brain. This investigation confirmed that intranasal delivery of oligomeric amyloid-β antibody (NU4) utilized all three routes to enter the brain with a resident time of 96 hours post single bolus intranasal administration, and showed evidence of perikaryal and parenchymal uptake of NU4 in 5XFAD mouse brain, confirming the intranasal route as a non-invasive and efficient way of delivering therapeutics to the brain. In addition, this study demonstrated that intranasal delivery of NU4 antibody lowered cerebral amyloid-β and improved spatial learning in 5XFAD mice.
Collapse
Affiliation(s)
- Chun Xiao
- Neuroscience Research, Jesse Brown VA Medical Center, Chicago, IL 60612, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Chapman CD, Frey WH, Craft S, Danielyan L, Hallschmid M, Schiöth HB, Benedict C. Intranasal treatment of central nervous system dysfunction in humans. Pharm Res 2012; 30:2475-84. [PMID: 23135822 PMCID: PMC3761088 DOI: 10.1007/s11095-012-0915-1] [Citation(s) in RCA: 209] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 10/12/2012] [Indexed: 01/30/2023]
Abstract
One of the most challenging problems facing modern medicine is how to deliver a given drug to a specific target at the exclusion of other regions. For example, a variety of compounds have beneficial effects within the central nervous system (CNS), but unwanted side effects in the periphery. For such compounds, traditional oral or intravenous drug delivery fails to provide benefit without cost. However, intranasal delivery is emerging as a noninvasive option for delivering drugs to the CNS with minimal peripheral exposure. Additionally, this method facilitates the delivery of large and/or charged therapeutics, which fail to effectively cross the blood-brain barrier (BBB). Thus, for a variety of growth factors, hormones, neuropeptides and therapeutics including insulin, oxytocin, orexin, and even stem cells, intranasal delivery is emerging as an efficient method of administration, and represents a promising therapeutic strategy for the treatment of diseases with CNS involvement, such as obesity, Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, depression, anxiety, autism spectrum disorders, seizures, drug addiction, eating disorders, and stroke.
Collapse
Affiliation(s)
- Colin D Chapman
- Department of Neuroscience, Uppsala University, Box 593, Husargatan 3, Uppsala, Sweden.
| | | | | | | | | | | | | |
Collapse
|
31
|
Lochhead JJ, Thorne RG. Intranasal delivery of biologics to the central nervous system. Adv Drug Deliv Rev 2012; 64:614-28. [PMID: 22119441 DOI: 10.1016/j.addr.2011.11.002] [Citation(s) in RCA: 803] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 11/03/2011] [Accepted: 11/07/2011] [Indexed: 12/28/2022]
Abstract
Treatment of central nervous system (CNS) diseases is very difficult due to the blood-brain barrier's (BBB) ability to severely restrict entry of all but small, non-polar compounds. Intranasal administration is a non-invasive method of drug delivery which may bypass the BBB to allow therapeutic substances direct access to the CNS. Intranasal delivery of large molecular weight biologics such as proteins, gene vectors, and stem cells is a potentially useful strategy to treat a variety of diseases/disorders of the CNS including stroke, Parkinson's disease, multiple sclerosis, Alzheimer's disease, epilepsy, and psychiatric disorders. Here we give an overview of relevant nasal anatomy and physiology and discuss the pathways and mechanisms likely involved in drug transport from the nasal epithelium to the CNS. Finally we review both pre-clinical and clinical studies involving intranasal delivery of biologics to the CNS.
Collapse
Affiliation(s)
- Jeffrey J Lochhead
- Pharmaceutical Sciences Division, University of Wisconsin-Madison School of Pharmacy, Madison, WI 53705, USA
| | | |
Collapse
|
32
|
Silvestroff L, Bartucci S, Pasquini J, Franco P. Cuprizone-induced demyelination in the rat cerebral cortex and thyroid hormone effects on cortical remyelination. Exp Neurol 2012; 235:357-67. [PMID: 22421533 DOI: 10.1016/j.expneurol.2012.02.018] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 01/25/2012] [Accepted: 02/29/2012] [Indexed: 12/19/2022]
Abstract
Multiple Sclerosis (MS) is an inflammatory demyelinating disease of the Central Nervous System which is characterized by multifocal demyelinated lesions dispersed throughout the brain. Although white matter lesions have been the most extensively studied, cortical demyelinaton lesions are also detected in MS brains. Cuprizone (CPZ)-induced demyelination in rodents has been widely used as a model for MS. Most of these studies focus on oligodendrocyte-rich structures, such as the corpus callosum (CC) and the cerebellar peduncles. However, it has been recently described that CPZ administration in mice also produces cortical demyelination, resembling some of the lesions found in MS patients. In this work we used CPZ-demyelinating model in Wistar rats to study demyelination in cortical forebrain areas. At the ultrastructural level, demyelination in the cortex was observed before detectable myelin loss in the subcortical white matter. During the course of CPZ intoxication Myelin Basic Protein immunodetection was decreased in cortical layers I-III due to a reduction in the number of cortical oligodendrocytes (OL). Oligodendroglial loss in CPZ-intoxicated rats correlated with an increase in the number of Glial Fibrillary Acidic Protein positive astrocytes and a shift in the location of Carbonic Anhydrase II from OL to astrocytes. After removal of CPZ from the diet, we evaluate intranasal Thyroid hormone (TH) effects on the progression of cortical lesions. As previously reported in the CC, TH treatment also accelerates remyelination rate in the cortex compared to rats undergoing spontaneous remyelination. Our results suggest that manipulation of TH levels could be considered as a strategy to promote remyelination process in the cortex and to prevent neuronal irreversible damage in patients suffering from MS.
Collapse
Affiliation(s)
- Lucas Silvestroff
- Departamento de Química Biológica e Instituto de Química y Fisicoquímica Biológicas (IQUIFIB-CONICET) Facultad de Farmacia y Bioquímica, UBA. Junín 956, CABA (C1113AAD), Buenos Aires, Argentina.
| | | | | | | |
Collapse
|
33
|
Martinez JA, Kasamatsu M, Rosales-Hernandez A, Hanson LR, Frey WH, Toth CC. Comparison of central versus peripheral delivery of pregabalin in neuropathic pain states. Mol Pain 2012; 8:3. [PMID: 22236461 PMCID: PMC3285045 DOI: 10.1186/1744-8069-8-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2011] [Accepted: 01/11/2012] [Indexed: 11/10/2022] Open
Abstract
Background Although pregabalin therapy is beneficial for neuropathic pain (NeP) by targeting the CaVα2δ-1 subunit, its site of action is uncertain. Direct targeting of the central nervous system may be beneficial for the avoidance of systemic side effects. Results We used intranasal, intrathecal, and near-nerve chamber forms of delivery of varying concentrations of pregabalin or saline delivered over 14 days in rat models of experimental diabetic peripheral neuropathy and spinal nerve ligation. As well, radiolabelled pregabalin was administered to determine localization with different deliveries. We evaluated tactile allodynia and thermal hyperalgesia at multiple time points, and then analyzed harvested nervous system tissues for molecular and immunohistochemical changes in CaVα2δ-1 protein expression. Both intrathecal and intranasal pregabalin administration at high concentrations relieved NeP behaviors, while near-nerve pregabalin delivery had no effect. NeP was associated with upregulation of CACNA2D1 mRNA and CaVα2δ-1 protein within peripheral nerve, dorsal root ganglia (DRG), and dorsal spinal cord, but not brain. Pregabalin's effect was limited to suppression of CaVα2δ-1 protein (but not CACNA2D1 mRNA) expression at the spinal dorsal horn in neuropathic pain states. Dorsal root ligation prevented CaVα2δ-1 protein trafficking anterograde from the dorsal root ganglia to the dorsal horn after neuropathic pain initiation. Conclusions Either intranasal or intrathecal pregabalin relieves neuropathic pain behaviours, perhaps due to pregabalin's effect upon anterograde CaVα2δ-1 protein trafficking from the DRG to the dorsal horn. Intranasal delivery of agents such as pregabalin may be an attractive alternative to systemic therapy for management of neuropathic pain states.
Collapse
Affiliation(s)
- Jose A Martinez
- Department of Clinical Neurosciences and the University of Calgary, Calgary, AB, Canada
| | | | | | | | | | | |
Collapse
|
34
|
Cellular Therapy for Ischemic Stroke. Transl Stroke Res 2012. [DOI: 10.1007/978-1-4419-9530-8_38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
35
|
Frisca F, Sabbadini RA, Goldshmit Y, Pébay A. Biological Effects of Lysophosphatidic Acid in the Nervous System. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY VOLUME 296 2012; 296:273-322. [DOI: 10.1016/b978-0-12-394307-1.00005-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
36
|
Hematopoietic Growth Factor Family for Stroke Drug Development. Transl Stroke Res 2012. [DOI: 10.1007/978-1-4419-9530-8_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|