1
|
Kelliny S, Zhou X, Bobrovskaya L. Alzheimer's Disease and Frontotemporal Dementia: A Review of Pathophysiology and Therapeutic Approaches. J Neurosci Res 2025; 103:e70046. [PMID: 40387258 PMCID: PMC12087441 DOI: 10.1002/jnr.70046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 04/01/2025] [Accepted: 05/02/2025] [Indexed: 05/20/2025]
Abstract
Alzheimer's disease (AD) is a devastating form of dementia, with the number of affected individuals rising sharply. The main hallmarks of the disease include amyloid-beta plaque deposits and neurofibrillary tangles consisting of hyperphosphorylated tau protein, besides other pathological features that contribute to the disease's complexity. The causes of sporadic AD are multifactorial and mostly age-related and involve risk factors such as diabetes and cardiovascular or cerebrovascular disorders. Frontotemporal dementia (FTD) is another type of dementia characterized by a spectrum of behaviors, memory, and motor abnormalities and associated with abnormal depositions of protein aggregation, including tau protein. Currently approved medications are symptomatic, and no disease-modifying therapy is available to halt the disease progression. Therefore, the development of multi-targeted therapeutic approaches could hold promise for the treatment of AD and other neurodegenerative disorders, including tauopathies. In this article, we will discuss the pathophysiology of AD and FTD, the proposed hypotheses, and current therapeutic approaches, highlighting the development of novel drug candidates and the progress of clinical trials in this field of research.
Collapse
Affiliation(s)
- Sally Kelliny
- Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
- Faculty of PharmacyAssiut UniversityAssiutEgypt
| | - Xin‐Fu Zhou
- Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Larisa Bobrovskaya
- Health and Biomedical Innovation, Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| |
Collapse
|
2
|
Ortega A, Chernicki B, Ou G, Parmar MS. From Lab Bench to Hope: Emerging Gene Therapies in Clinical Trials for Alzheimer's Disease. Mol Neurobiol 2025; 62:1112-1135. [PMID: 38958888 DOI: 10.1007/s12035-024-04285-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/05/2024] [Indexed: 07/04/2024]
Abstract
Alzheimer's disease is a progressive neurodegenerative disorder that affects memory and cognitive abilities, affecting millions of people around the world. Current treatments focus on the management of symptoms, as no effective therapy has been approved to modify the underlying disease process. Gene therapy is a promising approach that can offer disease-modifying treatment for AD, targeting various aspects of the pathophysiology of the disease. This review presents a comprehensive overview of the current state of gene therapy research for AD, with a specific focus on clinical trials and preclinical studies that have used nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), apolipoprotein E2 (APOE2), and human telomerase reverse transcriptase (hTERT) as therapeutic gene therapy approaches. These gene targets have shown potential to alleviate the neuropathology of AD in animal studies and have demonstrated feasibility and safety in non-human primates. Despite the failure of the NGF gene therapy approach in clinical trials, we have reviewed and highlighted the reported findings and evaluations from the trials. Furthermore, the review included the conclusions of postmortem brain tissue analysis of AD patients who received NGF gene therapy. The goal is to learn from the failed trials and improve the approach in the future. Although gene therapy shows promise, it faces several challenges and limitations, including optimizing gene delivery methods, enhancing safety and efficacy profiles, and determining long-term results. This review contributes to the growing body of literature on innovative treatments for AD and highlights the need for more research and development to advance gene therapy as a viable treatment option for AD.
Collapse
Affiliation(s)
- Angelica Ortega
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA
| | - Brendan Chernicki
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA
| | - Grace Ou
- College of Arts and Sciences, Cornell University, Ithaca, NY, USA
| | - Mayur S Parmar
- Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA.
- Department of Foundational Sciences, Dr. Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Clearwater, FL, USA.
| |
Collapse
|
3
|
Mitra S, Gera R, Eriksdotter M. NGF-based cholinergic therapies in Alzheimer disease. HANDBOOK OF CLINICAL NEUROLOGY 2025; 211:123-135. [PMID: 40340057 DOI: 10.1016/b978-0-443-19088-9.00007-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
The cholinergic system is part of the parasympathetic nervous system, which works in tandem with the sympathetic and enteric nervous systems to maintain the physiologic functioning of our body. The neurotransmitter acetylcholine (ACh) facilitates cholinergic signaling pathways by activating specific cell surface receptors (nicotinic and muscarinic receptors). Altered cholinergic signaling has been implicated in several pathologic conditions. In this chapter, conditions where cholinergic modulation in the central nervous system occurs through the neurotrophin nerve growth factor (NGF) are addressed. NGF is the master regulator of several pathways, ultimately leading to cell survival, ACh production, regenerative signaling, and anti-inflammatory tone. NGF and cholinergic-related pathways have been reported to be severely affected in the case of Alzheimer disease (AD), the most common dementia disorder. In AD, the cholinergic nuclei of the basal forebrain are affected early during the AD continuum, resulting in cholinergic cell loss and hampered ACh production, which overall affects the propagation of cholinergic signals in other brain regions. Since the 1990s clinically relevant strategies to treat AD patients have been the drugs that enhance cholinergic signaling-termed cholinesterase inhibitors (ChEIs), however, other strategies in AD have been and are presently being assessed for clinical efficacy. Delivery of NGF to the basal forebrain is considered crucial to revive the cholinergic cell bodies, restore ACh production, and sustain cognitive function. This chapter provides a description of the relevance of NGF-based therapies targeted for AD treatment, technical approaches for NGF delivery to the brain, and the status of ongoing clinical studies are provided.
Collapse
Affiliation(s)
- Sumonto Mitra
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Ruchi Gera
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Maria Eriksdotter
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden; Theme Inflammation and Aging, Medical Unit Aging, Karolinska University Hospital, Huddinge, Sweden.
| |
Collapse
|
4
|
Ebert ET, Schwinghamer KM, Siahaan TJ. Delivery of Neuroregenerative Proteins to the Brain for Treatments of Neurodegenerative Brain Diseases. Life (Basel) 2024; 14:1456. [PMID: 39598254 PMCID: PMC11595909 DOI: 10.3390/life14111456] [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: 09/21/2024] [Revised: 11/01/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
Neurodegenerative brain diseases such as Alzheimer's disease (AD), multiple sclerosis (MS), and Parkinson's disease (PD) are difficult to treat. Unfortunately, many therapeutic agents for neurodegenerative disease only halt the progression of these diseases and do not reverse neuronal damage. There is a demand for finding solutions to reverse neuronal damage in the central nervous system (CNS) of patients with neurodegenerative brain diseases. Therefore, the purpose of this review is to discuss the potential for therapeutic agents like specific neurotrophic and growth factors in promoting CNS neuroregeneration in brain diseases. We discuss how BDNF, NGF, IGF-1, and LIF could potentially be used for the treatment of brain diseases. The molecule's different mechanisms of action in stimulating neuroregeneration and methods to analyze their efficacy are described. Methods that can be utilized to deliver these proteins to the brain are also discussed.
Collapse
Affiliation(s)
| | | | - Teruna J. Siahaan
- Department of Pharmaceutical Chemistry, School of Pharmacy, The University of Kansas, 2095 Constant Avenue, Lawrence, KS 66047, USA; (E.T.E.); (K.M.S.)
| |
Collapse
|
5
|
Moskalenko AM, Ikrin AN, Kozlova AV, Mukhamadeev RR, de Abreu MS, Riga V, Kolesnikova TO, Kalueff AV. Decoding Molecular Bases of Rodent Social Hetero-Grooming Behavior Using in Silico Analyses and Bioinformatics Tools. Neuroscience 2024; 554:146-155. [PMID: 38876356 DOI: 10.1016/j.neuroscience.2024.06.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/03/2024] [Accepted: 06/06/2024] [Indexed: 06/16/2024]
Abstract
Highly prevalent in laboratory rodents, 'social' hetero-grooming behavior is translationally relevant to modeling a wide range of neuropsychiatric disorders. Here, we comprehensively evaluated all known to date mouse genes linked to aberrant hetero-grooming phenotype, and applied bioinformatics tools to construct a network of their established protein-protein interactions (PPI). We next identified several distinct molecular clusters within this complex network, including neuronal differentiation, cytoskeletal, WNT-signaling and synapsins-associated pathways. Using additional bioinformatics analyses, we further identified 'central' (hub) proteins within these molecular clusters, likely key for mouse hetero-grooming behavior. Overall, a more comprehensive characterization of intricate molecular pathways linked to aberrant rodent grooming may markedly advance our understanding of underlying cellular mechanisms and related neurological disorders, eventually helping discover novel targets for their pharmacological or gene therapy interventions.
Collapse
Affiliation(s)
- Anastasia M Moskalenko
- Graduate Program in Genetics and Genetic Technologies, Sirius University of Science and Technology, Sochi 354340, Russia; Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Aleksey N Ikrin
- Graduate Program in Genetics and Genetic Technologies, Sirius University of Science and Technology, Sochi 354340, Russia; Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Alena V Kozlova
- Graduate Program in Genetics and Genetic Technologies, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Radmir R Mukhamadeev
- Graduate Program in Bioinformatics and Genomics, Sirius University of Science and Technology, Sochi 354340, Russia; Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Murilo S de Abreu
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050, Brazil.
| | - Vyacheslav Riga
- Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Tatiana O Kolesnikova
- Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia
| | - Allan V Kalueff
- Neuroscience Department, Sirius University of Science and Technology, Sochi 354340, Russia; Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg 199034, Russia; Institute of Experimental Medicine, Almazov National Medical Research Centre, Ministry of Healthcare of Russian Federation, St. Petersburg 194021, Russia; Suzhou Key Laboratory of Neurobiology and Cell Signaling, Department of Biological Sciences, School of Science, Xi'an Jiaotong-Liverpool University (XJTLU), Suzhou 215123, China.
| |
Collapse
|
6
|
Tiberi A, Borgonovo G, Testa G, Pacifico P, Jacob A, Di Caprio M, Totaro V, Calvello M, Cattaneo A, Capsoni S. Reversal of neurological deficits by painless nerve growth factor in a mouse model of Rett syndrome. Brain 2024; 147:122-134. [PMID: 37633263 PMCID: PMC10766238 DOI: 10.1093/brain/awad282] [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: 01/23/2023] [Revised: 07/12/2023] [Accepted: 07/31/2023] [Indexed: 08/28/2023] Open
Abstract
Rett syndrome is a rare genetic neurodevelopmental disease, affecting 1 in over 10 000 females born worldwide, caused by de novo mutations in the X-chromosome-located methyl-CpG-binding protein 2 (MeCP2) gene. Despite the great effort put forth by the scientific community, a therapy for this devastating disease is still needed. Here, we tested the therapeutic effects of a painless mutein of the nerve growth factor (NGF), called human NGF painless (hNGFp), via a non-invasive intranasal delivery in female MeCP2+/- mice. Of note, previous work had demonstrated a broad biodistribution of hNGFp in the mouse brain by the nasal delivery route. We report that (i) the long-term lifelong treatment of MeCP2+/- mice with hNGFp, starting at 2 months of age, increased the chance of survival while also greatly improving behavioural parameters. Furthermore, when we assessed the phenotypic changes brought forth by (ii) a short-term 1-month-long hNGFp-treatment, starting at 3 months of age (right after the initial presentation of symptoms), we observed the rescue of a well known neuronal target population of NGF, cholinergic neurons in the medial septum. Moreover, we reveal a deficit in microglial morphology in MeCP2+/- mice, completely reversed in treated animals. This effect on microglia is in line with reports showing microglia to be a TrkA-dependent non-neuronal target cell population of NGF in the brain. To understand the immunomodulatory activity of hNGFp, we analysed the cytokine profile after hNGFp treatment in MeCP2+/- mice, to discover that the treatment recovered the altered expression of key neuroimmune-communication molecules, such as fractalkine. The overall conclusion is that hNGFp delivered intranasally can ameliorate symptoms in the MeCP2+/- model of Rett syndrome, by exerting strong neuroprotection with a dual mechanism of action: directly on target neurons and indirectly via microglia.
Collapse
Affiliation(s)
- Alexia Tiberi
- Institute of Neuroscience, CNR, 56124 Pisa, Italy
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
| | - Giulia Borgonovo
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
| | - Giovanna Testa
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
| | - Paola Pacifico
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
| | - Ajesh Jacob
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
| | | | - Valentino Totaro
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
| | | | - Antonino Cattaneo
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
- Rita Levi-Montalcini European Brain Research Institute (EBRI), 00161 Roma, Italy
| | - Simona Capsoni
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, 56126 Pisa, Italy
- Section of Human Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
7
|
Fronza MG, Alves D, Praticò D, Savegnago L. The neurobiology and therapeutic potential of multi-targeting β-secretase, glycogen synthase kinase 3β and acetylcholinesterase in Alzheimer's disease. Ageing Res Rev 2023; 90:102033. [PMID: 37595640 DOI: 10.1016/j.arr.2023.102033] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/04/2023] [Accepted: 08/14/2023] [Indexed: 08/20/2023]
Abstract
Alzheimer's Disease (AD) is the most common form of dementia, affecting almost 50 million of people around the world, characterized by a complex and age-related progressive pathology with projections to duplicate its incidence by the end of 2050. AD pathology has two major hallmarks, the amyloid beta (Aβ) peptides accumulation and tau hyperphosphorylation, alongside with several sub pathologies including neuroinflammation, oxidative stress, loss of neurogenesis and synaptic dysfunction. In recent years, extensive research pointed out several therapeutic targets which have shown promising effects on modifying the course of the disease in preclinical models of AD but with substantial failure when transposed to clinic trials, suggesting that modulating just an isolated feature of the pathology might not be sufficient to improve brain function and enhance cognition. In line with this, there is a growing consensus that an ideal disease modifying drug should address more than one feature of the pathology. Considering these evidence, β-secretase (BACE1), Glycogen synthase kinase 3β (GSK-3β) and acetylcholinesterase (AChE) has emerged as interesting therapeutic targets. BACE1 is the rate-limiting step in the Aβ production, GSK-3β is considered the main kinase responsible for Tau hyperphosphorylation, and AChE play an important role in modulating memory formation and learning. However, the effects underlying the modulation of these enzymes are not limited by its primarily functions, showing interesting effects in a wide range of impaired events secondary to AD pathology. In this sense, this review will summarize the involvement of BACE1, GSK-3β and AChE on synaptic function, neuroplasticity, neuroinflammation and oxidative stress. Additionally, we will present and discuss new perspectives on the modulation of these pathways on AD pathology and future directions on the development of drugs that concomitantly target these enzymes.
Collapse
Affiliation(s)
- Mariana G Fronza
- Neurobiotechnology Research Group (GPN) - Centre for Technology Development CDTec, Federal University of Pelotas (UFPel), Pelotas, RS, Brazil
| | - Diego Alves
- Laboratory of Clean Organic Synthesis (LASOL), Center for Chemical, Pharmaceutical and Food Sciences (CCQFA), UFPel, RS, Brazil
| | - Domenico Praticò
- Alzheimer's Center at Temple - ACT, Temple University, Lewis Katz School of Medicine, Philadelphia, PA, United States
| | - Lucielli Savegnago
- Neurobiotechnology Research Group (GPN) - Centre for Technology Development CDTec, Federal University of Pelotas (UFPel), Pelotas, RS, Brazil.
| |
Collapse
|
8
|
Pernet V, Joly S, Spiegel S, Meli I, Idriss S, Maigler F, Mdzomba JB, Roenneke AK, Franceschini A, Silvestri L, Pavone FS, Calamai M, Schindowski K, Chan A. Nogo-A antibody delivery through the olfactory mucosa mitigates experimental autoimmune encephalomyelitis in the mouse CNS. Cell Death Discov 2023; 9:290. [PMID: 37558696 PMCID: PMC10412545 DOI: 10.1038/s41420-023-01588-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 07/18/2023] [Accepted: 07/28/2023] [Indexed: 08/11/2023] Open
Abstract
Systemic administration of Nogo-A-neutralizing antibody ameliorates experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. However, the blood-brain barrier (BBB) is a major obstacle limiting the passage of systemically applied antibody to the CNS. To bypass the BBB, in the present study we tested the intranasal route of administration by targeting the olfactory mucosa with the Nogo-A-blocking antibody 11C7 mAb in myelin oligodendrocyte glycoprotein-induced EAE. Antibodies were specifically administered onto the olfactory mucosa using a microcatheter. Antibody distribution was examined in the CNS by ELISA and light-sheet microscopy. The effects of 11C7 mAb on Nogo-A signaling were assessed by Western blotting. EAE-induced deficits were monitored daily. Demyelination was observed on spinal cord histological sections. Gene expression changes were followed by trancriptomic analyses. A sensitive capture ELISA revealed a rapid and widespread distribution of 11C7 mAb in the CNS, including the olfactory bulb, the cerebellum and the lumbar spinal cord, but not in the CSF. Light-sheet microscopy allowed to observe antibody accumulation in the parenchyma, thus demonstrating nose-to-brain transfer of IgG. At the functional level, the widespread penetration of 11C7 mAb in the CNS, including the thoracolumbar spinal cord, resulted in the improvement of motor symptoms and in the preservation of myelin in the spinal cord of EAE mice. This was accompanied by Nogo-A signaling downregulation, as reflected by the decreased level of phosphorylated cofilin observed by Western blotting in the cerebellum. In the brain of EAE score-matched animals, 11C7 modified the expression of genes that can influence neurotransmission and cognitive functions, independently of the demyelination phenotype in the spinal cord. In conclusion, our data show the feasibility of olfactory mucosa-directed administration for the delivery of therapeutic antibodies targeting CNS antigens in EAE mice.
Collapse
Affiliation(s)
- Vincent Pernet
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Center for experimental neurology (ZEN), Bern University Hospital, University of Bern, Bern, Switzerland.
- Centre de recherche du CHU de Québec-Université Laval and Department of Molecular Medicine, Faculté de médecine, Université Laval, Québec, Québec, Canada.
| | - Sandrine Joly
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Center for experimental neurology (ZEN), Bern University Hospital, University of Bern, Bern, Switzerland
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Sebastian Spiegel
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Center for experimental neurology (ZEN), Bern University Hospital, University of Bern, Bern, Switzerland
- Institute of Applied Biotechnology, Biberach University of Applied Science, Hubertus-Liebrecht-Strasse 35, Biberach, Germany
- Department of Biomedical Research, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Ivo Meli
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Center for experimental neurology (ZEN), Bern University Hospital, University of Bern, Bern, Switzerland
| | - Sherif Idriss
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Center for experimental neurology (ZEN), Bern University Hospital, University of Bern, Bern, Switzerland
| | - Frank Maigler
- Institute of Applied Biotechnology, Biberach University of Applied Science, Hubertus-Liebrecht-Strasse 35, Biberach, Germany
| | - Julius Baya Mdzomba
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Center for experimental neurology (ZEN), Bern University Hospital, University of Bern, Bern, Switzerland
| | - Anna K Roenneke
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Center for experimental neurology (ZEN), Bern University Hospital, University of Bern, Bern, Switzerland
| | - Alessandra Franceschini
- LENS- European Laboratory for Non-Linear Spectroscopy, University of Florence, Sesto-Fiorentino (Firenze), Italy
| | - Ludovico Silvestri
- LENS- European Laboratory for Non-Linear Spectroscopy, University of Florence, Sesto-Fiorentino (Firenze), Italy
| | - Francesco S Pavone
- LENS- European Laboratory for Non-Linear Spectroscopy, University of Florence, Sesto-Fiorentino (Firenze), Italy
| | - Martino Calamai
- LENS- European Laboratory for Non-Linear Spectroscopy, University of Florence, Sesto-Fiorentino (Firenze), Italy
- National Institute of Optics - National Research Council (CNR-INO), Sesto Fiorentino, Italy
| | - Katharina Schindowski
- Institute of Applied Biotechnology, Biberach University of Applied Science, Hubertus-Liebrecht-Strasse 35, Biberach, Germany
| | - Andrew Chan
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Center for experimental neurology (ZEN), Bern University Hospital, University of Bern, Bern, Switzerland.
- Department of Biomedical Research, University of Bern, Bern, Switzerland.
| |
Collapse
|
9
|
Kim KH. Intranasal delivery of mitochondrial protein humanin rescues cell death and promotes mitochondrial function in Parkinson's disease. Theranostics 2023; 13:3330-3345. [PMID: 37351170 PMCID: PMC10283052 DOI: 10.7150/thno.84165] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/21/2023] [Indexed: 06/24/2023] Open
Abstract
Rationale: Mitochondrial dysfunction is a key factor in the pathogenesis of Parkinson's disease (PD). Accordingly, many aspects of mitochondrial function have been studied as a putative therapeutic target. Here we present a novel strategy to promote mitochondrial function and protect against Parkinson's disease by the peptide encoded within mitochondrial genome, mitochondria-derived peptide (MDP) humanin (HN). Methods: To test humanin as a potential biomarker in PD, we measured protein levels of circulating humanin from the plasma of PD patients and transgenic or neurotoxic mouse models of PD. Next, we aimed to identify whether HN peptide treatment can regulate its activity or expression. Using mouse models of PD, we assessed HN delivery to the brain via the nasal route of administration. We further revealed a possible mechanism underlying the therapeutic effectiveness of HN peptide for PD using in vitro and ex vivo model of PD. Results: Although the expression of intracellular HN was not correlated with PD, HN treatment itself could induce intracellular HN expression and enhance mitochondrial biogenesis inducing mitochondrial gene expression. After intranasal administration, HN peptide resulted in neuroprotection and behavioral recovery in an animal model of PD. Interestingly, HN peptide following intranasal delivery was found within the brain, mainly via the trigeminal pathways. Mechanistically, HN treatment induced activation of phosphatidylinositol-3-kinase/protein kinase B (PI3K/AKT) signaling pathway which led to enhanced mitochondrial biogenesis resulting in upregulation of mitochondrial gene including humanin. Conclusion: These data support a novel role of mitochondrial protein humanin in mitochondrial function and neuronal survival against Parkinson's disease, in which humanin treatment is sufficient for stimulating mitochondrial gene expression.
Collapse
Affiliation(s)
- Kyung Hwa Kim
- Department of Health Sciences, The Graduate School of Dong-A University, 840 Hadan-dong, Saha-gu, Busan 49315, Korea
| |
Collapse
|
10
|
Tiberi A, Carucci NM, Testa G, Rizzi C, Pacifico P, Borgonovo G, Arisi I, D’Onofrio M, Brandi R, Gan WB, Capsoni S, Cattaneo A. Reduced levels of NGF shift astrocytes toward a neurotoxic phenotype. Front Cell Dev Biol 2023; 11:1165125. [PMID: 37143894 PMCID: PMC10151754 DOI: 10.3389/fcell.2023.1165125] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 03/16/2023] [Indexed: 05/06/2023] Open
Abstract
Nerve growth factor (NGF) is critical for neuronal physiology during development and adulthood. Despite the well-recognized effect of NGF on neurons, less is known about whether NGF can actually affect other cell types in the central nervous system (CNS). In this work, we show that astrocytes are susceptible to changes in ambient levels of NGF. First, we observe that interfering with NGF signaling in vivo via the constitutive expression of an antiNGF antibody induces astrocytic atrophy. A similar asthenic phenotype is encountered in an uncleavable proNGF transgenic mouse model (TgproNGF#72), effectively increasing the brain proNGF levels. To examine whether this effect on astrocytes is cell-autonomous, we cultured wild-type primary astrocytes in the presence of antiNGF antibodies, uncovering that a short incubation period is sufficient to potently and rapidly trigger calcium oscillations. Acute induction of calcium oscillations by antiNGF antibodies is followed by progressive morphological changes similar to those observed in antiNGF AD11 mice. Conversely, incubation with mature NGF has no effect on either calcium activity nor on astrocytic morphology. At longer timescales, transcriptomic analysis revealed that NGF-deprived astrocytes acquire a proinflammatory profile. In particular, antiNGF-treated astrocytes show upregulation of neurotoxic transcripts and downregulation of neuroprotective mRNAs. Consistent with that data, culturing wild-type neurons in the presence of NGF-deprived astrocytes leads to neuronal cell death. Finally, we report that in both awake and anesthetized mice, astrocytes in layer I of the motor cortex respond with an increase in calcium activity to acute NGF inhibition using either NGF-neutralizing antibodies or a TrkA-Fc NGF scavenger. Moreover, in vivo calcium imaging in the cortex of the 5xFAD neurodegeneration mouse model shows an increased level of spontaneous calcium activity in astrocytes, which is significantly reduced after acute administration of NGF. In conclusion, we unveil a novel neurotoxic mechanism driven by astrocytes, triggered by their sensing and reacting to changes in the levels of ambient NGF.
Collapse
Affiliation(s)
- Alexia Tiberi
- BIO@SNS, Scuola Normale Superiore, Pisa, Italy
- Skirball Institute of Biomolecular Medicine, Langone Medical Center, New York University, New York, NY, United States
| | | | | | | | | | | | - Ivan Arisi
- European Brain Research Institute - Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Mara D’Onofrio
- European Brain Research Institute - Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Rossella Brandi
- European Brain Research Institute - Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Wen-Biao Gan
- Skirball Institute of Biomolecular Medicine, Langone Medical Center, New York University, New York, NY, United States
- Shenzhen Bay Laboratory, Shenzhen, China
| | - Simona Capsoni
- BIO@SNS, Scuola Normale Superiore, Pisa, Italy
- Institute of Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Antonino Cattaneo
- BIO@SNS, Scuola Normale Superiore, Pisa, Italy
- European Brain Research Institute - Fondazione Rita Levi-Montalcini, Rome, Italy
- *Correspondence: Antonino Cattaneo,
| |
Collapse
|
11
|
Current Strategies to Enhance Delivery of Drugs across the Blood–Brain Barrier. Pharmaceutics 2022; 14:pharmaceutics14050987. [PMID: 35631573 PMCID: PMC9145636 DOI: 10.3390/pharmaceutics14050987] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 04/18/2022] [Accepted: 04/29/2022] [Indexed: 12/13/2022] Open
Abstract
The blood–brain barrier (BBB) has shown to be a significant obstacle to brain medication delivery. The BBB in a healthy brain is a diffusion barrier that prevents most substances from passing from the blood to the brain; only tiny molecules can pass across the BBB. The BBB is disturbed in specific pathological illnesses such as stroke, diabetes, seizures, multiple sclerosis, Parkinson’s disease, and Alzheimer’s disease. The goal of this study is to offer a general overview of current brain medication delivery techniques and associated topics from the last five years. It is anticipated that this review will stimulate readers to look into new ways to deliver medications to the brain. Following an introduction of the construction and function of the BBB in both healthy and pathological conditions, this review revisits certain contested questions, such as whether nanoparticles may cross the BBB on their own and if medications are selectively delivered to the brain by deliberately targeted nanoparticles. Current non-nanoparticle options are also discussed, including drug delivery via the permeable BBB under pathological circumstances and the use of non-invasive approaches to improve brain medication absorption.
Collapse
|
12
|
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: 1.3] [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
|
13
|
Rogdakis T, Charou D, Latorrata A, Papadimitriou E, Tsengenes A, Athanasiou C, Papadopoulou M, Chalikiopoulou C, Katsila T, Ramos I, Prousis KC, Wade RC, Sidiropoulou K, Calogeropoulou T, Gravanis A, Charalampopoulos I. Development and Biological Characterization of a Novel Selective TrkA Agonist with Neuroprotective Properties against Amyloid Toxicity. Biomedicines 2022; 10:614. [PMID: 35327415 PMCID: PMC8945229 DOI: 10.3390/biomedicines10030614] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/23/2022] [Accepted: 03/03/2022] [Indexed: 12/12/2022] Open
Abstract
Neurotrophins are growth factors that exert important neuroprotective effects by preventing neuronal death and synaptic loss. Nerve Growth Factor (NGF) acts through the activation of its high-affinity, pro-survival TrkA and low-affinity, pro-apoptotic p75NTR receptors. NGF has been shown to slow or prevent neurodegenerative signals in Alzheimer's Disease (AD) progression. However, its low bioavailability and its blood-brain-barrier impermeability limit the use of NGF as a potential therapeutic agent against AD. Based on our previous findings on synthetic dehydroepiandrosterone derivatives, we identified a novel NGF mimetic, named ENT-A013, which selectively activates TrkA and exerts neuroprotective, anti-amyloid-β actions. We now report the chemical synthesis, in silico modelling, metabolic stability, CYP-mediated reaction phenotyping and biological characterization of ENT-A013 under physiological and neurodegenerative conditions. We show that ENT-A013 selectively activates the TrkA receptor and its downstream kinases Akt and Erk1/2 in PC12 cells, protecting these cells from serum deprivation-induced cell death. Moreover, ENT-A013 promotes survival of primary Dorsal Root Ganglion (DRG) neurons upon NGF withdrawal and protects hippocampal neurons against Amyloid β-induced apoptosis and synaptic loss. Furthermore, this neurotrophin mimetic partially restores LTP impairment. In conclusion, ENT-A013 represents a promising new lead molecule for developing therapeutics against neurodegenerative disorders, such as Alzheimer's Disease, selectively targeting TrkA-mediated pro-survival signals.
Collapse
Affiliation(s)
- Thanasis Rogdakis
- Department of Pharmacology, Medical School, University of Crete, 71003 Heraklion, Greece; (T.R.); (D.C.); (E.P.); (M.P.); (A.G.)
- Foundation for Research & Technology-Hellas (IMBB-FORTH), Institute of Molecular Biology & Biotechnology, 70013 Heraklion, Greece;
| | - Despoina Charou
- Department of Pharmacology, Medical School, University of Crete, 71003 Heraklion, Greece; (T.R.); (D.C.); (E.P.); (M.P.); (A.G.)
- Foundation for Research & Technology-Hellas (IMBB-FORTH), Institute of Molecular Biology & Biotechnology, 70013 Heraklion, Greece;
| | - Alessia Latorrata
- National Hellenic Research Foundation, Institute of Chemical Biology, 11635 Athens, Greece; (A.L.); (C.C.); (T.K.); (K.C.P.); (T.C.)
| | - Eleni Papadimitriou
- Department of Pharmacology, Medical School, University of Crete, 71003 Heraklion, Greece; (T.R.); (D.C.); (E.P.); (M.P.); (A.G.)
- Foundation for Research & Technology-Hellas (IMBB-FORTH), Institute of Molecular Biology & Biotechnology, 70013 Heraklion, Greece;
| | - Alexandros Tsengenes
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), 69118 Heidelberg, Germany; (A.T.); (C.A.); (R.C.W.)
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
- Heidelberg Biosciences International Graduate School, Heidelberg University, 69120 Heidelberg, Germany
| | - Christina Athanasiou
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), 69118 Heidelberg, Germany; (A.T.); (C.A.); (R.C.W.)
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
- Heidelberg Biosciences International Graduate School, Heidelberg University, 69120 Heidelberg, Germany
| | - Marianna Papadopoulou
- Department of Pharmacology, Medical School, University of Crete, 71003 Heraklion, Greece; (T.R.); (D.C.); (E.P.); (M.P.); (A.G.)
- Foundation for Research & Technology-Hellas (IMBB-FORTH), Institute of Molecular Biology & Biotechnology, 70013 Heraklion, Greece;
| | - Constantina Chalikiopoulou
- National Hellenic Research Foundation, Institute of Chemical Biology, 11635 Athens, Greece; (A.L.); (C.C.); (T.K.); (K.C.P.); (T.C.)
| | - Theodora Katsila
- National Hellenic Research Foundation, Institute of Chemical Biology, 11635 Athens, Greece; (A.L.); (C.C.); (T.K.); (K.C.P.); (T.C.)
| | - Isbaal Ramos
- Innovative Technologies in Biological Systems SL (INNOPROT), 48160 Bizkaia, Spain;
| | - Kyriakos C. Prousis
- National Hellenic Research Foundation, Institute of Chemical Biology, 11635 Athens, Greece; (A.L.); (C.C.); (T.K.); (K.C.P.); (T.C.)
| | - Rebecca C. Wade
- Molecular and Cellular Modeling Group, Heidelberg Institute for Theoretical Studies (HITS), 69118 Heidelberg, Germany; (A.T.); (C.A.); (R.C.W.)
- Faculty of Biosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Molecular Biology (ZMBH), DKFZ-ZMBH Alliance, Heidelberg University, 69120 Heidelberg, Germany
- Interdisciplinary Center for Scientific Computing (IWR), Heidelberg University, 69120 Heidelberg, Germany
| | - Kyriaki Sidiropoulou
- Foundation for Research & Technology-Hellas (IMBB-FORTH), Institute of Molecular Biology & Biotechnology, 70013 Heraklion, Greece;
- Department of Biology, University of Crete, 71113 Heraklion, Greece
| | - Theodora Calogeropoulou
- National Hellenic Research Foundation, Institute of Chemical Biology, 11635 Athens, Greece; (A.L.); (C.C.); (T.K.); (K.C.P.); (T.C.)
| | - Achille Gravanis
- Department of Pharmacology, Medical School, University of Crete, 71003 Heraklion, Greece; (T.R.); (D.C.); (E.P.); (M.P.); (A.G.)
- Foundation for Research & Technology-Hellas (IMBB-FORTH), Institute of Molecular Biology & Biotechnology, 70013 Heraklion, Greece;
| | - Ioannis Charalampopoulos
- Department of Pharmacology, Medical School, University of Crete, 71003 Heraklion, Greece; (T.R.); (D.C.); (E.P.); (M.P.); (A.G.)
- Foundation for Research & Technology-Hellas (IMBB-FORTH), Institute of Molecular Biology & Biotechnology, 70013 Heraklion, Greece;
| |
Collapse
|
14
|
Neuronal ApoE4 stimulates C/EBPβ activation, promoting Alzheimer’s disease pathology in a mouse model. Prog Neurobiol 2022; 209:102212. [DOI: 10.1016/j.pneurobio.2021.102212] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 12/05/2021] [Accepted: 12/22/2021] [Indexed: 12/15/2022]
|
15
|
Gao X, Liu N, Wang Z, Gao J, Zhang H, Li M, Du Y, Gao X, Zheng A. Development and Optimization of Chitosan Nanoparticle-Based Intranasal Vaccine Carrier. MOLECULES (BASEL, SWITZERLAND) 2021; 27:molecules27010204. [PMID: 35011436 PMCID: PMC8746444 DOI: 10.3390/molecules27010204] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/26/2021] [Accepted: 12/27/2021] [Indexed: 12/17/2022]
Abstract
Chitosan is a natural polysaccharide, mainly derived from the shell of marine organisms. At present, chitosan has been widely used in the field of biomedicine due to its special characteristics of low toxicity, biocompatibility, biodegradation and low immunogenicity. Chitosan nanoparticles can be easily prepared. Chitosan nanoparticles with positive charge can enhance the adhesion of antigens in nasal mucosa and promote its absorption, which is expected to be used for intranasal vaccine delivery. In this study, we prepared chitosan nanoparticles by a gelation method, and modified the chitosan nanoparticles with mannose by hybridization. Bovine serum albumin (BSA) was used as the model antigen for development of an intranasal vaccine. The preparation technology of the chitosan nanoparticle-based intranasal vaccine delivery system was optimized by design of experiment (DoE). The DoE results showed that mannose-modified chitosan nanoparticles (Man-BSA-CS-NPs) had high modification tolerance and the mean particle size and the surface charge with optimized Man-BSA-CS-NPs were 156 nm and +33.5 mV. FTIR and DSC results confirmed the presence of Man in Man-BSA-CS-NPs. The BSA released from Man-BSA-CS-NPs had no irreversible aggregation or degradation. In addition, the analysis of fluorescence spectroscopy of BSA confirmed an appropriate binding constant between CS and BSA in this study, which could improve the stability of BSA. The cell study in vitro demonstrated the low toxicity and biocompatibility of Man-BSA-CS-NPs. Confocal results showed that the Man-modified BSA-FITC-CS-NPs promote the endocytosis and internalization of BSA-FITC in DC2.4 cells. In vivo studies of mice, Man-BSA-CS-NPs intranasally immunized showed a significantly improvement of BSA-specific serum IgG response and the highest level of BSA-specific IgA expression in nasal lavage fluid. Overall, our study provides a promising method to modify BSA-loaded CS-NPs with mannose, which is worthy of further study.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yimeng Du
- Correspondence: (Y.D.); (X.G.); (A.Z.); Tel.: +86-010-6693-1694 or +86-135-2046-7936 (A.Z.)
| | - Xiang Gao
- Correspondence: (Y.D.); (X.G.); (A.Z.); Tel.: +86-010-6693-1694 or +86-135-2046-7936 (A.Z.)
| | - Aiping Zheng
- Correspondence: (Y.D.); (X.G.); (A.Z.); Tel.: +86-010-6693-1694 or +86-135-2046-7936 (A.Z.)
| |
Collapse
|
16
|
Manni L, Conti G, Chiaretti A, Soligo M. Intranasal Delivery of Nerve Growth Factor in Neurodegenerative Diseases and Neurotrauma. Front Pharmacol 2021; 12:754502. [PMID: 34867367 PMCID: PMC8635100 DOI: 10.3389/fphar.2021.754502] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/01/2021] [Indexed: 01/01/2023] Open
Abstract
Since the 1980s, the development of a pharmacology based on nerve growth factor (NGF) has been postulated for the therapy of Alzheimer’s disease (AD). This hypothesis was based on the rescuing effect of the neurotrophin on the cholinergic phenotype of the basal forebrain neurons, primarily compromised during the development of AD. Subsequently, the use of NGF was put forward to treat a broader spectrum of neurological conditions affecting the central nervous system, such as Parkinson’s disease, degenerative retinopathies, severe brain traumas and neurodevelopmental dysfunctions. While supported by solid rational assumptions, the progress of a pharmacology founded on these hypotheses has been hampered by the difficulty of conveying NGF towards the brain parenchyma without resorting to invasive and risky delivery methods. At the end of the last century, it was shown that NGF administered intranasally to the olfactory epithelium was able to spread into the brain parenchyma. Notably, after such delivery, pharmacologically relevant concentration of exogenous NGF was found in brain areas located at considerable distances from the injection site along the rostral-caudal axis. These observations paved the way for preclinical characterization and clinical trials on the efficacy of intranasal NGF for the treatment of neurodegenerative diseases and of the consequences of brain trauma. In this review, a summary of the preclinical and clinical studies published to date will be attempted, as well as a discussion about the mechanisms underlying the efficacy and the possible development of the pharmacology based on intranasal conveyance of NGF to the brain.
Collapse
Affiliation(s)
- Luigi Manni
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Rome, Italy
| | - Giorgio Conti
- Department of Emergency, Intensive Pediatric Therapy and Pediatric Trauma Center, Anesthesiological and Reanimation Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Antonio Chiaretti
- Department of Woman and Child Health, Institute of Pediatrics, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Marzia Soligo
- Institute of Translational Pharmacology, National Research Council of Italy (CNR), Rome, Italy
| |
Collapse
|
17
|
Metz CN, Pavlov VA. Treating disorders across the lifespan by modulating cholinergic signaling with galantamine. J Neurochem 2021; 158:1359-1380. [PMID: 33219523 PMCID: PMC10049459 DOI: 10.1111/jnc.15243] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/15/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023]
Abstract
Advances in understanding the regulatory functions of the nervous system have revealed neural cholinergic signaling as a key regulator of cytokine responses and inflammation. Cholinergic drugs, including the centrally acting acetylcholinesterase inhibitor, galantamine, which are in clinical use for the treatment of Alzheimer's disease and other neurodegenerative and neuropsychiatric disorders, have been rediscovered as anti-inflammatory agents. Here, we provide a timely update on this active research and clinical developments. We summarize the involvement of cholinergic mechanisms and inflammation in the pathobiology of Alzheimer's disease, Parkinson's disease, and schizophrenia, and the effectiveness of galantamine treatment. We also highlight recent findings demonstrating the effects of galantamine in preclinical and clinical settings of numerous conditions and diseases across the lifespan that are characterized by immunological, neurological, and metabolic dysfunction.
Collapse
Affiliation(s)
- Christine N. Metz
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Valentin A. Pavlov
- The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| |
Collapse
|
18
|
Mitra S, Gera R, Linderoth B, Lind G, Wahlberg L, Almqvist P, Behbahani H, Eriksdotter M. A Review of Techniques for Biodelivery of Nerve Growth Factor (NGF) to the Brain in Relation to Alzheimer's Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1331:167-191. [PMID: 34453298 DOI: 10.1007/978-3-030-74046-7_11] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Age-dependent progressive neurodegeneration and associated cognitive dysfunction represent a serious concern worldwide. Currently, dementia accounts for the fifth highest cause of death, among which Alzheimer's disease (AD) represents more than 60% of the cases. AD is associated with progressive cognitive dysfunction which affects daily life of the affected individual and associated family. The cognitive dysfunctions are at least partially due to the degeneration of a specific set of neurons (cholinergic neurons) whose cell bodies are situated in the basal forebrain region (basal forebrain cholinergic neurons, BFCNs) but innervate wide areas of the brain. It has been explicitly shown that the delivery of the neurotrophic protein nerve growth factor (NGF) can rescue BFCNs and restore cognitive dysfunction, making NGF interesting as a potential therapeutic substance for AD. Unfortunately, NGF cannot pass through the blood-brain barrier (BBB) and thus peripheral administration of NGF protein is not viable therapeutically. NGF must be delivered in a way which will allow its brain penetration and availability to the BFCNs to modulate BFCN activity and viability. Over the past few decades, various methodologies have been developed to deliver NGF to the brain tissue. In this chapter, NGF delivery methods are discussed in the context of AD.
Collapse
Affiliation(s)
- Sumonto Mitra
- Division of Clinical Geriatrics, NVS Department, Karolinska Institutet, Stockholm, Sweden.
| | - Ruchi Gera
- Division of Clinical Geriatrics, NVS Department, Karolinska Institutet, Stockholm, Sweden
| | - Bengt Linderoth
- Section of Neurosurgery, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Göran Lind
- Section of Neurosurgery, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Per Almqvist
- Section of Neurosurgery, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Homira Behbahani
- Division of Clinical Geriatrics, NVS Department, Karolinska Institutet, Stockholm, Sweden.,Karolinska Universitets laboratoriet (LNP5), Karolinska University Hospital, Stockholm, Sweden
| | - Maria Eriksdotter
- Division of Clinical Geriatrics, NVS Department, Karolinska Institutet, Stockholm, Sweden.,Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
19
|
Mladenova K, Stavrakov G, Philipova I, Atanasova M, Petrova S, Doumanov J, Doytchinova I. A Galantamine-Curcumin Hybrid Decreases the Cytotoxicity of Amyloid-Beta Peptide on SH-SY5Y Cells. Int J Mol Sci 2021; 22:7592. [PMID: 34299209 PMCID: PMC8307467 DOI: 10.3390/ijms22147592] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 11/18/2022] Open
Abstract
Misfolded amyloid beta (Aβ) peptides aggregate and form neurotoxic oligomers. Membrane and mitochondrial damages, calcium dysregulation, oxidative stress, and fibril deposits are among the possible mechanisms of Aβ cytotoxicity. Galantamine (GAL) prevents apoptosis induced by Aβ mainly through the ability to stimulate allosterically the α7 nAChRs and to regulate the calcium cytosolic concentration. Here, we examined the cytoprotective effects of two GAL derivatives, namely compounds 4b and 8, against Aβ cytotoxicity on the human neuroblastoma cell line SH-SY5Y. The protective effects were tested at simultaneous administration, pre-incubation and post-incubation, with Aβ. GAL and curcumin (CU) were used in the study as reference compounds. It was found that 4b protects cells in a similar mode as GAL, while compound 8 and CU potentiate the toxic effects of Aβ. Allosteric stimulation of α7 nAChRs is suggested as a possible mechanism of the cytoprotectivity of 4b. These and previous findings characterize 4b as a prospective non-toxic multi-target agent against neurodegenerative disorders with inhibitory activity on acetylcholinesterase, antioxidant, and cytoprotective properties.
Collapse
Affiliation(s)
- Kirilka Mladenova
- Faculty of Biology, Sofia University, 1164 Sofia, Bulgaria; (K.M.); (S.P.); (J.D.)
| | - Georgi Stavrakov
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (G.S.); (M.A.)
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;
| | - Irena Philipova
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;
| | - Mariyana Atanasova
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (G.S.); (M.A.)
| | - Svetla Petrova
- Faculty of Biology, Sofia University, 1164 Sofia, Bulgaria; (K.M.); (S.P.); (J.D.)
| | - Jordan Doumanov
- Faculty of Biology, Sofia University, 1164 Sofia, Bulgaria; (K.M.); (S.P.); (J.D.)
| | - Irini Doytchinova
- Faculty of Pharmacy, Medical University of Sofia, 1000 Sofia, Bulgaria; (G.S.); (M.A.)
| |
Collapse
|
20
|
Vazquez-Roque R, Pacheco-Flores M, Penagos-Corzo JC, Flores G, Aguilera J, Treviño S, Guevara J, Diaz A, Venegas B. The C-terminal fragment of the heavy chain of the tetanus toxin (Hc-TeTx) improves motor activity and neuronal morphology in the limbic system of aged mice. Synapse 2021; 75:e22193. [PMID: 33141999 DOI: 10.1002/syn.22193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 10/24/2020] [Accepted: 10/27/2020] [Indexed: 12/23/2022]
Abstract
In the aging process, the brain presents biochemical and morphological alterations. The neurons of the limbic system show reduced size dendrites, in addition to the loss of dendritic spines. These disturbances trigger a decrease in motor and cognitive function. Likewise, it is reported that during aging, in the brain, there is a significant decrease in neurotrophic factors, which are essential in promoting the survival and plasticity of neurons. The carboxyl-terminal fragment of the heavy chain of the tetanus toxin (Hc-TeTx) acts similarly to neurotrophic factors, inducing neuroprotection in different models of neuronal damage. The aim here, was to evaluate the effect of Hc-TeTx on the motor processes of elderly mice (18 months old), and its impact on the dendritic morphology and density of dendritic spines of neurons in the limbic system. The morphological analysis in the dendrites was evaluated employing Golgi-Cox staining. Hc-TeTx was administered (0.5 mg/kg) intraperitoneally for three days in 18-month-old mice. Locomotor activity was evaluated in a novel environment 30 days after the last administration of Hc-TeTx. Mice treated with Hc-TeTx showed significant changes in their motor behavior, and an increased dendritic spine density of pyramidal neurons in layers 3 and 5 of the prefrontal cortex in the hippocampus, and medium spiny neurons of the nucleus accumbens (NAcc). In conclusion, the Hc-TeTx improves the plasticity of the brain regions of the limbic system of aged mice. Therefore, it is proposed as a pharmacological alternative to prevent or delay brain damage during aging.
Collapse
Affiliation(s)
- Ruben Vazquez-Roque
- Neuropsychiatry Laboratory, Institute of Physiology, Benemérita Universidad Autónoma de Puebla, Puebla, México
| | | | | | - Gonzalo Flores
- Neuropsychiatry Laboratory, Institute of Physiology, Benemérita Universidad Autónoma de Puebla, Puebla, México
| | - José Aguilera
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Institut de Neurociències, Universitat Autònoma de Barcelona, Cerdanyola del Vallès, Barcelona, Spain
- Networked Biomedical Research Center on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Samuel Treviño
- Faculty of Chemical Sciences, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Jorge Guevara
- Department of Biochemistry, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| | - Alfonso Diaz
- Faculty of Chemical Sciences, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Berenice Venegas
- Faculty of Biological Sciences, Benemérita Universidad Autónoma de Puebla, Puebla, México
| |
Collapse
|
21
|
Braschi C, Capsoni S, Narducci R, Poli A, Sansevero G, Brandi R, Maffei L, Cattaneo A, Berardi N. Intranasal delivery of BDNF rescues memory deficits in AD11 mice and reduces brain microgliosis. Aging Clin Exp Res 2021; 33:1223-1238. [PMID: 32676979 PMCID: PMC8081712 DOI: 10.1007/s40520-020-01646-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 06/29/2020] [Indexed: 01/22/2023]
Abstract
A decrease in brain-derived neurotrophic factor (BDNF), a neurotrophin essential for synaptic function, plasticity and neuronal survival, is evident early in the progression of Alzheimer's disease (AD), being apparent in subjects with mild cognitive impairment or mild AD, and both proBDNF and mature BDNF levels are positively correlated with cognitive measures. BDNF delivery is, therefore, considered of great interest as a potentially useful therapeutic strategy to contrast AD. Invasive BDNF administration has indeed been recently used in animal models of AD with promising results in rescuing memory deficits, synaptic density and cell loss. Here, we tested whether non-invasive intranasal administration of different BDNF concentrations after the onset of cognitive and anatomical deficits (6 months of age) could rescue neuropathological and memory deficits in AD11 mice, a model of NGF deprivation-induced neurodegeneration. In addition to AD hallmarks, we investigated BDNF effects on microglia presence in the brain of AD11 mice, since alterations in microglia activation have been associated with ageing-related cognitive decline and with the progression of neurodegenerative diseases, including AD. We found that intranasal delivery of 42 pmol BDNF (1 μM), but not PBS, was sufficient to completely rescue performance of AD11 mice both in the object recognition test and in the object context test. No further improvement was obtained with 420 pmol (10 μM) BDNF dose. The strong improvement in memory performance in BDNF-treated mice was not accompanied by an amelioration of AD-like pathology, Aβ burden, tau hyperphosphorylation and cholinergic deficit, but there was a dramatic decrease of CD11b immunoreactive brain microglia. These results reinforce the potential therapeutic uses of BDNF in AD and the non-invasive intranasal route as an effective delivery strategy of BDNF to the brain. They also strengthen the connection between neuroinflammation and neurodegenerative dementia and suggest microglia as a possible mediator of BDNF therapeutic actions in the brain.
Collapse
Affiliation(s)
- Chiara Braschi
- Institute of Neuroscience of the CNR, Via G. Moruzzi 1, 56124, Pisa, Italy
- Department of Neuroscience, Psychology, Drug Research, Child Health (NEUROFARBA), Florence University, Florence, Italy
| | - Simona Capsoni
- Scuola Normale Superiore, Pisa, Italy
- Human Physiology Section, Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Roberta Narducci
- Institute of Neuroscience of the CNR, Via G. Moruzzi 1, 56124, Pisa, Italy
- Department of Neuroscience, Psychology, Drug Research, Child Health (NEUROFARBA), Florence University, Florence, Italy
| | | | - Gabriele Sansevero
- Institute of Neuroscience of the CNR, Via G. Moruzzi 1, 56124, Pisa, Italy
- IRCCS Stella Maris, Calambrone, Pisa, Italy
| | | | - Lamberto Maffei
- Institute of Neuroscience of the CNR, Via G. Moruzzi 1, 56124, Pisa, Italy
- Scuola Normale Superiore, Pisa, Italy
| | - Antonino Cattaneo
- Scuola Normale Superiore, Pisa, Italy
- European Brain Research Institute, Rome, Italy
| | - Nicoletta Berardi
- Institute of Neuroscience of the CNR, Via G. Moruzzi 1, 56124, Pisa, Italy.
- Department of Neuroscience, Psychology, Drug Research, Child Health (NEUROFARBA), Florence University, Florence, Italy.
| |
Collapse
|
22
|
Luo J, Yang Y, Ji X, He W, Fan J, Huang Y, Wang Y. NGF Rescues Spermatogenesis in Azoospermic Mice. Reprod Sci 2021; 28:2780-2788. [PMID: 33725311 DOI: 10.1007/s43032-021-00511-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 02/21/2021] [Indexed: 11/26/2022]
Abstract
Nerve growth factor (NGF) plays an important role in regulating the hypothalamus-pituitary-gonadal (HPG) axis. However, the effects of NGF on spermatogenesis remain unclear. This study aimed to assess the potential application of NGF with nasal delivery on spermatogenesis in azoospermic mice. We established a model with azoospermia induced by a single intraperitoneal (i.p.) injection of busulfan. NGF pre-encapsulated with liposomes (25, 50, and 100 μg/kg) was delivered via internasal administration. Three weeks after busulfan injection, NGF treatments were performed twice a week for 8 weeks; the change of sperm quality, testis and epididymis histopathology, and androgenic hormone were analyzed to evaluate sperm regeneration. Furthermore, 30 mg/kg busulfan injection caused severe testicular atrophy of the seminiferous tubules, characterized by a loss of spermatogenic elements and sperms. NGF with nasal administration could significantly upregulate the markers expressing meiotic spermatogonia (Stra8) and spermatocytes (SYCP3), restore spermatogenesis, and improve sperm quality in busulfan-treated mice by increasing the secretion of sexual hormones. The convenient and noninvasive nasal delivery of NGF may be a new potential therapy for spermatogenesis via activating the HPG axis and elevating androgenic hormones. This study opened a new horizon for NGF application in reproductive endocrine.
Collapse
Affiliation(s)
- Jiao Luo
- Department of Rehabilitation Medicine, Dapeng New District Nan'ao People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Department of Neurology, The First Affiliated Hospital of Shenzhen University, Institute for Translational Medicine, Shenzhen Second People's Hospital, Shenzhen, China
- Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences, Shenzhen, China
| | - Yan Yang
- Department of Cell Biology, Jinan University, Guangzhou, China
| | - Xunmin Ji
- Guangdong Provincial Institute of Biological Products and Materia, Guangzhou, China
| | - Weiyi He
- School of Public Health, Health Science Center, Shenzhen University, Shenzhen, China
| | - Jing Fan
- Department of Reproductive Medicine Center, Shenzhen Maternity and Child Healthcare Hospital, Shenzhen, Guangdong Province, China
| | - Yadong Huang
- Department of Cell Biology, Jinan University, Guangzhou, China
- Department of Pharmacology, Jinan University, Guangzhou, China
| | - Yulong Wang
- Department of Rehabilitation, Shenzhen Second People's Hospital, The First Affiliated Hospital, Shenzhen University School of Medicine, Shenzhen, China.
| |
Collapse
|
23
|
Wang ZH, Xia Y, Liu P, Liu X, Edgington-Mitchell L, Lei K, Yu SP, Wang XC, Ye K. ApoE4 activates C/EBPβ/δ-secretase with 27-hydroxycholesterol, driving the pathogenesis of Alzheimer's disease. Prog Neurobiol 2021; 202:102032. [PMID: 33716161 DOI: 10.1016/j.pneurobio.2021.102032] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/17/2021] [Accepted: 03/09/2021] [Indexed: 02/06/2023]
Abstract
ApoE4, an apolipoprotein implicated in cholesterol transport and amyloid-β (Aβ) metabolism, is a major genetic risk determinant for Alzheimer's Disease (AD) and drives its pathogenesis via Aβ-dependent and -independent pathways. C/EBPβ, a proinflammatory cytokines-activated transcription factor, is upregulated in AD and mediates cytokines and δ-secretase expression. However, how ApoE4 contributes to AD pathogenesis remains incompletely understood. Here we show that ApoE4 and 27-hydroxycholesterol (27-OHC) co-activate C/EBPβ/δ-secretase signaling in neurons, mediating AD pathogenesis, and this effect is dependent on neuronal secreted Aβ and inflammatory cytokines. Inhibition of cholesterol metabolism with lovastatin diminishes neuronal ApoE4's stimulatory effects. Furthermore, ApoE4 and 27-OHC also mediate lysosomal δ-secretase leakage, activation, secretion and endocytosis. Notably, 27-OHC strongly activates C/EBPβ/δ-secretase pathway in human ApoE4-TR mice and triggers AD pathologies and cognitive deficits, which is blocked by C/EBPβ depletion. Hence, our findings demonstrate that ApoE4 and 27-OHC additively trigger AD pathogenesis via activating C/EBPβ/δ-secretase pathway. Lowering cholesterol levels with statins should benefit the ApoE4 AD carriers.
Collapse
Affiliation(s)
- Zhi-Hao Wang
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA; Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yiyuan Xia
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA; Department of Pathophysiology, Key Laboratory of Ministry of Education of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pai Liu
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA; Neuroscience Graduate Program, Laney Graduate School, Emory University, Atlanta, GA, 30322, USA
| | - Xia Liu
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Laura Edgington-Mitchell
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne Victoria, 3010, Australia
| | - Kecheng Lei
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Shan Ping Yu
- Department of Anesthesiology, Emory University School of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Xiao-Chuan Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education of Neurological Diseases, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, JS, 226001, China.
| | - Keqiang Ye
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
24
|
Kanu LN, Ciolino JB. Nerve Growth Factor as an Ocular Therapy: Applications, Challenges, and Future Directions. Semin Ophthalmol 2021; 36:224-231. [PMID: 33641595 DOI: 10.1080/08820538.2021.1890793] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Nerve growth factor (NGF), the prototypical neurotrophin first discovered in the 1950s, has recently garnered increased interest as a therapeutic agent promoting neuronal health and regeneration. After gaining orphan drug status within the last decade, NGF-related research and drug development has accelerated. The purpose of this article is to review the preclinical and clinical evidence of NGF in various applications, including central and peripheral nervous system, skin, and ophthalmic disorders. We focus on the ophthalmic applications including not only the FDA-approved indication of neurotrophic keratitis but also retinal disease and glaucoma. NGF represents a promising therapy whose therapeutic profile is evolving. The challenges related to this therapy are reviewed, along with possible solutions and future directions.
Collapse
Affiliation(s)
- Levi N Kanu
- 1. Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Joseph B Ciolino
- 1. Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
25
|
Ye D, Luan J, Pang H, Yang Y, Nazeri A, Rubin JB, Chen H. Characterization of focused ultrasound-mediated brainstem delivery of intranasally administered agents. J Control Release 2020; 328:276-285. [PMID: 32871204 PMCID: PMC7749082 DOI: 10.1016/j.jconrel.2020.08.053] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 08/22/2020] [Accepted: 08/25/2020] [Indexed: 12/14/2022]
Abstract
Focused ultrasound-mediated intranasal (FUSIN) delivery is a recently proposed technique that bypasses the blood-brain barrier to achieve noninvasive and localized brain drug delivery. The goal of this study was to characterize FUSIN drug delivery outcome in mice with regard to its dependency on several critical experimental factors, including the time interval between IN administration and FUS sonication (Tlag1), the FUS pressure, and the time for sacrificing the mice post-FUS (Tlag2). Wild-type mice were treated by FUSIN delivery of near-infrared fluorescent dye-labeled bovine serum albumin (800CW-BSA, used as a model agent). 800CW-BSA was intranasally administered to the mice in vivo, followed by intravenous injection of microbubbles and FUS sonication at the brainstem. Fluorescence imaging of ex vivo mouse brain slices was used to quantify the delivery outcomes of 800CW-BSA. Major organs, along with the nasal tissue and trigeminal nerve, were harvested to assess the biodistribution of 800CW-BSA. The delivery outcome of 800CW-BSA was the highest at the brainstem when Tlag1 was 0.5 h, which was on average 24.5-fold, 5.4-fold, and 21.6-fold higher than those of the IN only, Tlag1 = 1.5 h, and Tlag1 = 4.0 h, respectively. The FUSIN delivery outcome at the lowest pressure level, 0.43 MPa, was on average 1.8-fold and 3.7-fold higher than those at 0.56 MPa and 0.70 MPa, respectively. The mean concentration of 800CW-BSA in the brainstem after FUSIN delivery decreased from 0.5 h to 4.0 h post-FUS. The accumulation of 800CW-BSA was low in the heart, lung, spleen, kidneys, and liver, but high in the stomach and intestines. This study revealed the unique characteristics of FUSIN as a noninvasive, efficient, and localized brain drug delivery technique.
Collapse
Affiliation(s)
- Dezhuang Ye
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, Saint Louis, MO, 63130, USA
| | - Jingyi Luan
- Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, Saint Louis, MO, 63130, USA
| | - Hannah Pang
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, 63130, USA
| | - Yaoheng Yang
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, 63130, USA
| | - Arash Nazeri
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO., 63110, USA
| | - Joshua B Rubin
- Departments of Pediatrics and Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO, 63130, USA; Department of Radiation Oncology, Washington University School of Medicine, Saint Louis, MO, 63108, USA..
| |
Collapse
|
26
|
Kuo YC, Lou YI, Rajesh R, Chen CL. Multiple-component dual-phase solid lipid nanoparticles with conjugated transferrin for formulating antioxidants and nerve growth factor against neuronal apoptosis. J Taiwan Inst Chem Eng 2020. [DOI: 10.1016/j.jtice.2020.02.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
27
|
Padmakumar S, Taha MS, Kadakia E, Bleier BS, Amiji MM. Delivery of neurotrophic factors in the treatment of age-related chronic neurodegenerative diseases. Expert Opin Drug Deliv 2020; 17:323-340. [DOI: 10.1080/17425247.2020.1727443] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Smrithi Padmakumar
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, USA
| | - Maie S. Taha
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, USA
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ekta Kadakia
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, USA
- Drug Metabolism and Pharmacokinetics (DMPK), Biogen Inc, Cambridge, MA, USA
| | - Benjamin S. Bleier
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, USA
| | - Mansoor M. Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy, Northeastern University, Boston, MA, USA
| |
Collapse
|
28
|
Zilony-Hanin N, Rosenberg M, Richman M, Yehuda R, Schori H, Motiei M, Rahimipour S, Groisman A, Segal E, Shefi O. Neuroprotective Effect of Nerve Growth Factor Loaded in Porous Silicon Nanostructures in an Alzheimer's Disease Model and Potential Delivery to the Brain. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2019; 15:e1904203. [PMID: 31482695 DOI: 10.1002/smll.201904203] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Indexed: 06/10/2023]
Abstract
Nerve growth factor (NGF) plays a vital role in reducing the loss of cholinergic neurons in Alzheimer's disease (AD). However, its delivery to the brain remains a challenge. Herein, NGF is loaded into degradable oxidized porous silicon (PSiO2 ) carriers, which are designed to carry and continuously release the protein over a 1 month period. The released NGF exhibits a substantial neuroprotective effect in differentiated rat pheochromocytoma PC12 cells against amyloid-beta (Aβ)-induced cytotoxicity, which is associated with Alzheimer's disease. Next, two potential localized administration routes of the porous carriers into murine brain are investigated: implantation of PSiO2 chips above the dura mater, and biolistic bombardment of PSiO2 microparticles through an opening in the skull using a pneumatic gene gun. The PSiO2 -implanted mice are monitored for a period of 8 weeks and no inflammation or adverse effects are observed. Subsequently, a successful biolistic delivery of these highly porous microparticles into a live-mouse brain is demonstrated for the first time. The bombarded microparticles are observed to penetrate the brain and reach a depth of 150 µm. These results pave the way for using degradable PSiO2 carriers as potential localized delivery systems for NGF to the brain.
Collapse
Affiliation(s)
- Neta Zilony-Hanin
- Faculty of Engineering, Bar-Ilan University, Ramat-Gan, 5290002, Israel
- Bar-Ilan Institute of Nanotechnologies and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Michal Rosenberg
- Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Michal Richman
- Department of Chemistry, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Ronen Yehuda
- Department of Life Sciences, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Hadas Schori
- Faculty of Engineering, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Menachem Motiei
- Faculty of Engineering, Bar-Ilan University, Ramat-Gan, 5290002, Israel
- Bar-Ilan Institute of Nanotechnologies and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Shai Rahimipour
- Department of Chemistry, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| | - Alexander Groisman
- Department of Physics, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Ester Segal
- Department of Biotechnology and Food Engineering, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
- Russell Berrie Nanotechnology Institute, Technion - Israel Institute of Technology, Haifa, 3200003, Israel
| | - Orit Shefi
- Faculty of Engineering, Bar-Ilan University, Ramat-Gan, 5290002, Israel
- Bar-Ilan Institute of Nanotechnologies and Advanced Materials, Bar-Ilan University, Ramat-Gan, 5290002, Israel
| |
Collapse
|
29
|
Amadoro G, Latina V, Corsetti V, Calissano P. N-terminal tau truncation in the pathogenesis of Alzheimer's disease (AD): Developing a novel diagnostic and therapeutic approach. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165584. [PMID: 31676377 DOI: 10.1016/j.bbadis.2019.165584] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 01/04/2023]
Abstract
Tau truncation occurs at early stages during the development of human Alzheimer's disease (AD) and other tauopathy dementias. Tau cleavage, particularly in its N-terminal projection domain, is able to drive per se neurodegeneration, regardless of its pro-aggregative pathway(s) and in fragment(s)-dependent way. In this short review, we highlight the pathological relevance of the 20-22 kDa NH2-truncated tau fragment which is endowed with potent neurotoxic "gain-of-function" action(s), both in vitro and in vivo. An extensive comment on its clinical value as novel progression/diagnostic biomarker and potential therapeutic target in the context of tau-mediated neurodegeneration is also provided.
Collapse
Affiliation(s)
- G Amadoro
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy; Institute of Translational Pharmacology (IFT)-CNR, Via Fosso del Cavaliere 100, 00133 Rome, Italy.
| | - V Latina
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - V Corsetti
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - P Calissano
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| |
Collapse
|
30
|
Zhao D, Gu MY, Zhang LJ, Jeon HJ, Cho YB, Yang HO. 7-Deoxy- trans-dihydronarciclasine Isolated from Lycoris chejuensis Inhibits Neuroinflammation in Experimental Models. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:9796-9804. [PMID: 31393712 DOI: 10.1021/acs.jafc.9b03307] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Overactivated microglia and persistent neuroinflammation hold an important role in the pathophysiology of neurodegenerative diseases. The extract of Lycoris chejuensis (CJ) and its active compound, 7-deoxy-trans-dihydronarciclasine (named E144), attenuated expressions of pro-inflammatory factors, including nitric oxide, prostaglandin E2, inducible nitric oxide synthase, cyclooxygenase-2 (COX-2), tumor necrosis factor α (TNF-α), and interleukin 6, secreted by lipopolysaccharide-activated BV-2 microglial cells, as measured by an enzyme-linked immunosorbent assay or western blotting. In contrast, CJ extract and E144 promoted the secretion of the anti-inflammatory cytokine, interleukin 10. Moreover, we found that E144 attenuated the expression of TNF-α and COX-2 in the cerebral cortex of lipopolysaccharide-treated mice and/or T2576 transgenic mice as well as reduced the reactive immune cells visualized by ionized calcium-binding adaptor molecule 1. Our results suggest the possibility of E144 to serve as a potential anti-neuroinflammatory agent by preventing excess production of pro-inflammatory factors.
Collapse
Affiliation(s)
- Dong Zhao
- Natural Product Research Center , Korea Institute of Science and Technology , Gangneung , Gangwon-do 25451 , Republic of Korea
- Division of Bio-Medical Science & Technology, Korea Institute of Science and Technology (KIST) School , Korea University of Science and Technology , Seoul 02792 , Republic of Korea
| | - Ming-Yao Gu
- Department of Cell Biology and Medical Genetics, School of Basic Medical Sciences , Shenzhen University Health Science Center , Shenzhen , Guangdong 51801 , People's Republic of China
| | - Li Jun Zhang
- Natural Product Research Center , Korea Institute of Science and Technology , Gangneung , Gangwon-do 25451 , Republic of Korea
- Division of Bio-Medical Science & Technology, Korea Institute of Science and Technology (KIST) School , Korea University of Science and Technology , Seoul 02792 , Republic of Korea
| | - Hyo Jin Jeon
- Pharmaceutical R&D Center , Kolmar Korea , Sejong 30003 , Republic of Korea
| | - Yong-Baik Cho
- Pharmaceutical R&D Center , Kolmar Korea , Sejong 30003 , Republic of Korea
| | - Hyun Ok Yang
- Natural Product Research Center , Korea Institute of Science and Technology , Gangneung , Gangwon-do 25451 , Republic of Korea
- Division of Bio-Medical Science & Technology, Korea Institute of Science and Technology (KIST) School , Korea University of Science and Technology , Seoul 02792 , Republic of Korea
| |
Collapse
|
31
|
Xiong L, Duan L, Xu W, Wang Z. Nerve growth factor metabolic dysfunction contributes to sevoflurane-induced cholinergic degeneration and cognitive impairments. Brain Res 2018; 1707:107-116. [PMID: 30481505 DOI: 10.1016/j.brainres.2018.11.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 11/21/2018] [Accepted: 11/24/2018] [Indexed: 12/26/2022]
Abstract
General anesthesia with sevoflurane is associated with an increased incidence of postoperative cognitive dysfunction. Previous studies have shown that sevoflurane anesthesia can affect the integrity and function of basal forebrain cholinergic neurons (BFCNs) which are essential for learning and memory. However, the underlying mechanisms remain largely unknown. Here, we demonstrated that exposure to 2.5% sevoflurane induced significant loss of BFCNs and caused impairments of the spatial and the fear memory. Further, sevoflurane exposure significantly reduced the level of nerve growth factor (NGF), an important factor for the survival and phenotype maintenance of BFCNs, by disrupting its synthesis pathways in the brain. More importantly, NGF administration not only prevented the loss of BFCNs but also ameliorated the cognitive impairments in sevoflurane-treated mice. Our findings indicate that NGF metabolic dysfunction contributes to sevoflurane-associated BFCNs degeneration and subsequent cognitive deficits.
Collapse
Affiliation(s)
- Lu Xiong
- Department of Anesthesiology, Tinglin Hospital of Jinshan Disctrict, Shanghai 201505, China
| | - Lijie Duan
- Department of Neurology, Jinshan Hospital, Fudan University, Shanghai 201508, China
| | - Wenqing Xu
- Department of Anesthesiology, Tinglin Hospital of Jinshan Disctrict, Shanghai 201505, China
| | - Zigao Wang
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
32
|
Cattaneo A, Capsoni S. Painless Nerve Growth Factor: A TrkA biased agonist mediating a broad neuroprotection via its actions on microglia cells. Pharmacol Res 2018; 139:17-25. [PMID: 30391352 DOI: 10.1016/j.phrs.2018.10.028] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/28/2018] [Accepted: 10/28/2018] [Indexed: 12/13/2022]
Abstract
Nerve Growth Factor (NGF) is a therapeutic candidate for Alzheimer's disease, based on its well known actions on basal forebrain cholinergic neurons. However, because of its pro-nociceptive activity, in current clinical trials NGF has to be administered intraparenchymally into the brain by neurosurgery via cell or gene therapy approaches. To prevent the NGF pain-inducing collateral effects, thus avoiding the necessity for local brain injection, we developed painless NGF (hNGFp), based on the human genetic disease Hereditary Sensory and Autonomic Neuropathy type V (HSAN V). hNGFp has similar neurotrophic activity as wild type human NGF, but its pain sensitizing activity is tenfold lower. Pharmacologically, hNGFp is a biased receptor agonist of NGF TrkA receptor. The results of recent studies shed new light on the neuroprotective mechanism by hNGFp and are highly relevant for the planning of NGF-based clinical trials. The intraparenchymal delivery of hNGFp, as used in clinical trials, was simulated in the 5xFAD mouse model and found to be inefficacious in reducing Aβ plaque load. On the contrary, the same dose of hNGFp administered intranasally, which was rather widely biodistributed in the brain and did not induce pain sensitization, blocked APP processing into amyloid and restored synaptic plasticity and memory in this aggressive neurodegeneration model. This potent and broad neuroprotection by hNGFp was found to be mediated by hNGFp actions on glial cells. hNGFp increases inflammatory proteins such as the soluble TNFα receptor II and the chemokine CXCL12. Independent work has shown that NGF has a potent anti-inflammatory action on microglia and steers them towards a neuroprotective phenotype. These studies demonstrate that microglia cells are a new target cell of NGF in the brain and have therapeutic significance: i) they establish that the neuroprotective actions of hNGFp relies on a widespread exposure of the brain, ii) they identify a new anti-neurodegenerative pathway, linking hNGFp to inflammatory chemokines and cytokines via microglia, a common target for new therapeutic opportunities for neurodegenerative diseases, iii) they extend the neuroprotective potential of hNGFp beyond its classical cholinergic target, thereby widening the range of neurological diseases for which this neurotrophic factor might be used therapeutically, iv) they help interpreting the results of current NGF clinical trials in AD and the design of future trials with this new potent therapeutic candidate.
Collapse
Affiliation(s)
- Antonino Cattaneo
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy; Rita Levi-Montalcini European Brain Research Institute (EBRI), Roma, Italy.
| | - Simona Capsoni
- Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy; Section of Human Physiology, Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| |
Collapse
|
33
|
Evaluation of intranasal delivery route of drug administration for brain targeting. Brain Res Bull 2018; 143:155-170. [PMID: 30449731 DOI: 10.1016/j.brainresbull.2018.10.009] [Citation(s) in RCA: 454] [Impact Index Per Article: 64.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/20/2018] [Accepted: 10/23/2018] [Indexed: 12/22/2022]
Abstract
The acute or chronic drug treatments for different neurodegenerative and psychiatric disorders are challenging from several aspects. The low bioavailability and limited brain exposure of oral drugs, the rapid metabolism, elimination, the unwanted side effects and also the high dose to be added mean both inconvenience for the patients and high costs for the patients, their family and the society. The reason of low brain penetration of the compounds is that they have to overcome the blood-brain barrier which protects the brain against xenobiotics. Intranasal drug administration is one of the promising options to bypass blood-brain barrier, to reduce the systemic adverse effects of the drugs and to lower the doses to be administered. Furthermore, the drugs administered using nasal route have usually higher bioavailability, less side effects and result in higher brain exposure at similar dosage than the oral drugs. In this review the focus is on giving an overview on the anatomical and cellular structure of nasal cavity and absorption surface. It presents some possibilities to enhance the drug penetration through the nasal barrier and summarizes some in vitro, ex vivo and in vivo technologies to test the drug delivery across the nasal epithelium into the brain. Finally, the authors give a critical evaluation of the nasal route of administration showing its main advantages and limitations of this delivery route for CNS drug targeting.
Collapse
|
34
|
Pham GS, Wang LA, Mathis KW. Pharmacological potentiation of the efferent vagus nerve attenuates blood pressure and renal injury in a murine model of systemic lupus erythematosus. Am J Physiol Regul Integr Comp Physiol 2018; 315:R1261-R1271. [PMID: 30332305 DOI: 10.1152/ajpregu.00362.2017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Recent evidence suggests hypertension may be secondary to chronic inflammation that results from hypoactive neuro-immune regulatory mechanisms. To further understand this association, we used systemic lupus erythematosus (SLE) as a model of inflammation-induced hypertension. In addition to prevalent inflammatory kidney disease and hypertension, SLE patients suffer from dysautonomia in the form of decreased efferent vagal tone. Based on this, the cholinergic anti-inflammatory pathway, an endogenous vagus-to-spleen mechanism that, when activated results in decreases in systemic inflammation, may be compromised in SLE. We hypothesized that stimulation of the cholinergic anti-inflammatory pathway via pharmacological potentiation of the efferent vagus nerve would reduce inflammation and halt the development of hypertension and renal injury in SLE. Female NZBWF1 mice, an established model of murine SLE, and female control mice were treated with galantamine (4 mg/kg daily ip), an acetylcholinesterase inhibitor, or saline for 14 days. At the end of therapy, carotid catheters were surgically implanted and were used to measure mean arterial pressure before the animals were euthanized. Chronic galantamine administration attenuated both splenic and renal cortical inflammation, which likely explains why the hypertension and renal injury (i.e., glomerulosclerosis and fibrosis) typically observed in murine SLE was attenuated following therapy. Based on this, the anti-inflammatory, antihypertensive, and renoprotective effects of galantamine may be mediated through activation of the cholinergic anti-inflammatory pathway. It is possible that dysfunction of the cholinergic anti-inflammatory pathway exists in SLE at the level of the efferent vagus nerve and promoting restoration of its activity through central cholinergic receptor activation may be beneficial.
Collapse
Affiliation(s)
- Grace S Pham
- Department of Physiology and Anatomy, University of North Texas Health Science Center , Fort Worth, Texas
| | - Lei A Wang
- Department of Physiology and Anatomy, University of North Texas Health Science Center , Fort Worth, Texas
| | - Keisa W Mathis
- Department of Physiology and Anatomy, University of North Texas Health Science Center , Fort Worth, Texas
| |
Collapse
|
35
|
Luo J, Yang Y, Zhang T, Su Z, Yu D, Lin Q, Chen H, Zhang Q, Xiang Q, Xue W, Ge R, Huang Y. Nasal delivery of nerve growth factor rescue hypogonadism by up-regulating GnRH and testosterone in aging male mice. EBioMedicine 2018; 35:295-306. [PMID: 30131307 PMCID: PMC6161474 DOI: 10.1016/j.ebiom.2018.08.021] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 08/08/2018] [Accepted: 08/08/2018] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Nerve growth factor (NGF) plays essential roles in regulating the development and maintenance of central sympathetic and sensory neurons. However, the effects of NGF on hypogonadism remain unexplored. METHODS To assess the effects of NGF on hypogonadism, we established a convenient and noninvasive way to deliver NGF to the hypothalamus by spraying liposome-encapsulated NGF into the nasal cavity. The ten-month-old aging male senescence accelerate mouse P8 (SAMP8) mice with age-related hypogonadotrophic hypogonadism were used to study the role of NGF in hypogonadism. The age-matched accelerated senescence-resistant mouse R1 (SAMR1) served as a control. The ten-month-old SAMP8 mice were treated with NGF twice per week for 12 weeks. Sexual hormones, sexual behaviors, and fertility were analyzed after NGF treatment. And the mechanisms of NGF in sex hormones sexual function were also studied. FINDINGS NGF could enhance the sexual function, improve the quality of the sperm, and restore the fertility of aging male SAMP8 mice with age-related hypogonadism by activating gonadotropin-releasing hormone (GnRH) neuron and regulating secretion of GnRH. And NGF regulated the GnRH release through the PKC/p-ERK1/2/p-CREB signal pathway. INTERPRETATION These results suggest that NGF treatment could alleviate various age-related hypogonadism symptoms in male SAMP8 and may be usefulness for age-related hypogonadotrophic hypogonadism and its related subfertility. FUND: National Natural Science Foundation of China, Natural Science Foundation of Guangdong Province, the Science and Technology Plan Project of Guangzhou, Wenzhou Science & Technology Bureau, Guangdong Province Pearl River Scholar Fund, Guangdong province science and technology innovation leading Scholar Fund.
Collapse
Affiliation(s)
- Jiao Luo
- Department of Cell Biology & Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Yan Yang
- Department of Cell Biology & Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China; Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Tiantian Zhang
- Department of Cell Biology & Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Zhijian Su
- Department of Cell Biology & Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Dan Yu
- Department of Pharmacology, Jinan University, Guangzhou 510632, China
| | - Qilian Lin
- Department of Cell Biology & Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Haolin Chen
- Center of Scientific Research, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Qihao Zhang
- Department of Cell Biology & Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Qi Xiang
- Department of Cell Biology & Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China; Department of Pharmacology, Jinan University, Guangzhou 510632, China
| | - Wei Xue
- Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Renshan Ge
- Center of Scientific Research, The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Yadong Huang
- Department of Cell Biology & Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China; Department of Pharmacology, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
36
|
The ProNGF/p75NTR pathway induces tau pathology and is a therapeutic target for FTLD-tau. Mol Psychiatry 2018; 23:1813-1824. [PMID: 29867188 DOI: 10.1038/s41380-018-0071-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 01/26/2018] [Accepted: 03/26/2018] [Indexed: 11/09/2022]
Abstract
Tau pathology is characterized as a form of frontotemporal lobar degeneration (FTLD) known as FTLD-tau. The underlying pathogenic mechanisms are not known and no therapeutic interventions are currently available. Here, we report that the neurotrophin receptor p75NTR plays a critical role in the pathogenesis of FTLD-tau. The expression of p75NTR and the precursor of nerve growth factor (proNGF) were increased in the brains of FTLD-tau patients and mice (P301L transgenic). ProNGF-induced tau phosphorylation via p75NTR in vitro, which was associated with the AKT/glycogen synthase kinase (GSK)3β pathway. Genetic reduction of p75NTR in P301L mice rescued the memory deficits, alleviated tau hyperphosphorylation and restored the activity of the AKT/GSK3β pathway. Treatment of the P301L mice with the soluble p75NTR extracellular domain (p75ECD-Fc), which can antagonize neurotoxic ligands of p75NTR, effectively improved memory behavior and suppressed tau pathology. This suggests that p75NTR plays a crucial role in tau paGSKthology and represents a potential druggable target for FTLD-tau and related tauopathies.
Collapse
|
37
|
Cruz E, Kumar S, Yuan L, Arikkath J, Batra SK. Intracellular amyloid beta expression leads to dysregulation of the mitogen-activated protein kinase and bone morphogenetic protein-2 signaling axis. PLoS One 2018; 13:e0191696. [PMID: 29470488 PMCID: PMC5823380 DOI: 10.1371/journal.pone.0191696] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/09/2018] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative syndrome classically depicted by the parenchymal accumulation of extracellular amyloid beta plaques. However, recent findings suggest intraneuronal amyloid beta (iAβ1-42) accumulation precedes extracellular deposition. Furthermore, the pathologic increase in iAβ1-42 has been implicated in dysregulation of cellular mechanisms critically important in axonal transport. Owing to neuronal cell polarity, retrograde and anterograde axonal transport are essential trafficking mechanism necessary to convey membrane bound neurotransmitters, neurotrophins, and endosomes between soma and synaptic interfaces. Although iAβ1-42 disruption of axonal transport has been implicated in dysregulation of neuronal synaptic transmission, the role of iAβ1-42 and its influence on signal transduction involving the mitogen-activated protein kinase (MAPK) and morphogenetic signaling axis are unknown. Our biochemical characterization of intracellular amyloid beta accumulation on MAPK and morphogenetic signaling have revealed increased iAβ1-42 expression leads to significant reduction in ERK 1/2 phosphorylation and increased bone morphogenetic protein 2 dependent Smad 1/5/8 phosphorylation. Furthermore, rescue of iAβ1-42 mediated attenuation of MAPK signaling can be accomplished with the small molecule PLX4032 as a downstream enhancer of the MAPK pathway. Consequently, our observations regarding the dysregulation of these gatekeepers of neuronal viability may have important implications in understanding the iAβ1-42 mediated effects observed in AD.
Collapse
Affiliation(s)
- Eric Cruz
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Li Yuan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Jyothi Arikkath
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Developmental Neuroscience, Munroe-Meyer Institute, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Surinder K. Batra
- Department of Biochemistry and Molecular Biology, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
| |
Collapse
|
38
|
Capsoni S, Malerba F, Carucci NM, Rizzi C, Criscuolo C, Origlia N, Calvello M, Viegi A, Meli G, Cattaneo A. The chemokine CXCL12 mediates the anti-amyloidogenic action of painless human nerve growth factor. Brain 2017; 140:201-217. [PMID: 28031222 PMCID: PMC5379860 DOI: 10.1093/brain/aww271] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 08/19/2016] [Accepted: 09/07/2016] [Indexed: 11/17/2022] Open
Abstract
Nerve growth factor is a therapeutic candidate for Alzheimer’s disease. Due to its pain-inducing activity, in current clinical trials nerve growth factor is delivered locally into the brain by neurosurgery, but data on the efficacy of local nerve growth factor delivery in decreasing amyloid-β deposition are not available. To reduce the nerve growth factor pain-inducing side effects, thus avoiding the need for local brain injection, we developed human painless nerve growth factor (hNGFp), inspired by the human genetic disease hereditary sensory and autonomic neuropathy type V. hNGFp has identical neurotrophic potency as wild-type human nerve growth factor, but a 10-fold lower pain sensitizing activity. In this study we first mimicked, in the 5xFAD mouse model, the intraparenchymal delivery of hNGFp used in clinical trials and found it to be ineffective in decreasing amyloid-β plaque load. On the contrary, the same dose of hNGFp delivered intranasally, which was widely biodistributed in the brain and did not induce pain, showed a potent anti-amyloidogenic action and rescued synaptic plasticity and memory deficits. We found that hNGFp acts on glial cells, modulating inflammatory proteins such as the soluble TNFα receptor II and the chemokine CXCL12. We further established that the rescuing effect by hNGFp is mediated by CXCL12, as pharmacological inhibition of CXCL12 receptor CXCR4 occludes most of hNGFp effects. These findings have significant therapeutic implications: (i) we established that a widespread exposure of the brain is required for nerve growth factor to fully exert its neuroprotective actions; and (ii) we have identified a new anti-neurodegenerative pathway as a broad target for new therapeutic opportunities for neurodegenerative diseases.
Collapse
Affiliation(s)
- Simona Capsoni
- 1 Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy.,2 Institute of Neuroscience, National Council for Research, Pisa, Italy
| | - Francesca Malerba
- 1 Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy.,3 Neurotrophins and Neurodegenerative Diseases Laboratory, Rita Levi-Montalcini European Brain Research Institute, Rome, Italy
| | | | - Caterina Rizzi
- 1 Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
| | - Chiara Criscuolo
- 2 Institute of Neuroscience, National Council for Research, Pisa, Italy.,4 Department of Biotechnological and Applied Clinical Sciences, School of Medicine, University of L'Aquila, Coppito, L'Aquila, Italy
| | - Nicola Origlia
- 2 Institute of Neuroscience, National Council for Research, Pisa, Italy
| | | | - Alessandro Viegi
- 1 Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy
| | - Giovanni Meli
- 3 Neurotrophins and Neurodegenerative Diseases Laboratory, Rita Levi-Montalcini European Brain Research Institute, Rome, Italy
| | - Antonino Cattaneo
- 1 Bio@SNS Laboratory of Biology, Scuola Normale Superiore, Pisa, Italy .,3 Neurotrophins and Neurodegenerative Diseases Laboratory, Rita Levi-Montalcini European Brain Research Institute, Rome, Italy
| |
Collapse
|
39
|
Nanotechnological strategies for nerve growth factor delivery: Therapeutic implications in Alzheimer’s disease. Pharmacol Res 2017; 120:68-87. [DOI: 10.1016/j.phrs.2017.03.020] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2016] [Revised: 02/23/2017] [Accepted: 03/22/2017] [Indexed: 12/30/2022]
|
40
|
Meng T, Cao Q, Lei P, Bush AI, Xiang Q, Su Z, He X, Rogers JT, Chiu IM, Zhang Q, Huang Y. Tat-haFGF 14-154 Upregulates ADAM10 to Attenuate the Alzheimer Phenotype of APP/PS1 Mice through the PI3K-CREB-IRE1α/XBP1 Pathway. MOLECULAR THERAPY-NUCLEIC ACIDS 2017. [PMID: 28624220 PMCID: PMC5443968 DOI: 10.1016/j.omtn.2017.05.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Acid fibroblast growth factor (aFGF) has shown neuroprotection in Alzheimer’s disease (AD) models in previous studies, yet its mechanism is still uncertain. Here we report that the efficacy of Tat-haFGF14–154 is markedly increased when loaded cationic liposomes for intranasal delivery are intranasally administered to APP/PS1 mice. Our results demonstrated that liposomal Tat-haFGF14–154 treatment significantly ameliorated behavioral deficits, relieved brain Aβ burden, and increased the expression and activity of disintegrin and metalloproteinase domain-containing protein 10 (ADAM10) in the brain. Tat-haFGF14–154 antagonized Aβ1–42-induced cell death and structural damage in rat primary neurons in an ADAM10-dependent manner, which, in turn, was promoted by the activation of XBP1 splicing and modulated by the PI3K-CREB pathway. Both knockdown of ADAM10 and inhibition of PI3K (LY294002) negated Tat-haFGF14–154 rescue. Thus, Tat-haFGF14–154 activates the IRE1α/XBP1 pathway of the unfolded protein response (UPR) against the endoplasmic reticulum (ER) stress induced by Aβ, and, subsequently, the nuclear translocation of spliced XBP1 (XBP1s) promotes transcription of ADAM10. These results highlight the important role of ADAM10 and its activation through the PI3K-CREB-IRE1α/XBP1 pathway as a key factor in the mechanism of neuroprotection for Tat-haFGF14–154.
Collapse
Affiliation(s)
- Tian Meng
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Qin Cao
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Peng Lei
- Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Ashley I Bush
- Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Qi Xiang
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China; Cell Biology Department and National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou 510632, China
| | - Zhijian Su
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China; Cell Biology Department and National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou 510632, China
| | - Xiang He
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Jack T Rogers
- Neurochemistry Laboratory, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02114, USA
| | - Ing-Ming Chiu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli 35053, Taiwan
| | - Qihao Zhang
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China; Cell Biology Department and National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou 510632, China.
| | - Yadong Huang
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China; Cell Biology Department and National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
41
|
Guo T, Noble W, Hanger DP. Roles of tau protein in health and disease. Acta Neuropathol 2017; 133:665-704. [PMID: 28386764 PMCID: PMC5390006 DOI: 10.1007/s00401-017-1707-9] [Citation(s) in RCA: 630] [Impact Index Per Article: 78.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/26/2017] [Accepted: 03/26/2017] [Indexed: 01/18/2023]
Abstract
Tau is well established as a microtubule-associated protein in neurons. However, under pathological conditions, aberrant assembly of tau into insoluble aggregates is accompanied by synaptic dysfunction and neural cell death in a range of neurodegenerative disorders, collectively referred to as tauopathies. Recent advances in our understanding of the multiple functions and different locations of tau inside and outside neurons have revealed novel insights into its importance in a diverse range of molecular pathways including cell signalling, synaptic plasticity, and regulation of genomic stability. The present review describes the physiological and pathophysiological properties of tau and how these relate to its distribution and functions in neurons. We highlight the post-translational modifications of tau, which are pivotal in defining and modulating tau localisation and its roles in health and disease. We include discussion of other pathologically relevant changes in tau, including mutation and aggregation, and how these aspects impinge on the propensity of tau to propagate, and potentially drive neuronal loss, in diseased brain. Finally, we describe the cascade of pathological events that may be driven by tau dysfunction, including impaired axonal transport, alterations in synapse and mitochondrial function, activation of the unfolded protein response and defective protein degradation. It is important to fully understand the range of neuronal functions attributed to tau, since this will provide vital information on its involvement in the development and pathogenesis of disease. Such knowledge will enable determination of which critical molecular pathways should be targeted by potential therapeutic agents developed for the treatment of tauopathies.
Collapse
Affiliation(s)
- Tong Guo
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 9NU, UK
| | - Wendy Noble
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 9NU, UK
| | - Diane P Hanger
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE5 9NU, UK.
| |
Collapse
|
42
|
Latina V, Caioli S, Zona C, Ciotti MT, Amadoro G, Calissano P. Impaired NGF/TrkA Signaling Causes Early AD-Linked Presynaptic Dysfunction in Cholinergic Primary Neurons. Front Cell Neurosci 2017; 11:68. [PMID: 28360840 PMCID: PMC5350152 DOI: 10.3389/fncel.2017.00068] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 02/24/2017] [Indexed: 12/31/2022] Open
Abstract
Alterations in NGF/TrkA signaling have been suggested to underlie the selective degeneration of the cholinergic basal forebrain neurons occurring in vivo in AD (Counts and Mufson, 2005; Mufson et al., 2008; Niewiadomska et al., 2011) and significant reduction of cognitive decline along with an improvement of cholinergic hypofunction have been found in phase I clinical trial in humans affected from mild AD following therapeutic NGF gene therapy (Tuszynski et al., 2005, 2015). Here, we show that the chronic (10–12 D.I.V.) in vitro treatment with NGF (100 ng/ml) under conditions of low supplementation (0.2%) with the culturing serum-substitute B27 selectively enriches the basal forebrain cholinergic neurons (+36.36%) at the expense of other non-cholinergic, mainly GABAergic (−38.45%) and glutamatergic (−56.25%), populations. By taking advantage of this newly-developed septo-hippocampal neuronal cultures, our biochemical and electrophysiological investigations demonstrate that the early failure in excitatory neurotransmission following NGF withdrawal is paralleled by concomitant and progressive loss in selected presynaptic and vesicles trafficking proteins including synapsin I, SNAP-25 and α-synuclein. This rapid presynaptic dysfunction: (i) precedes the commitment to cell death and is reversible in a time-dependent manner, being suppressed by de novo external administration of NGF within 6 hr from its initial withdrawal; (ii) is specific because it is not accompanied by contextual changes in expression levels of non-synaptic proteins from other subcellular compartments; (ii) is not secondary to axonal degeneration because it is insensible to pharmacological treatment with known microtubule-stabilizing drug such paclitaxel; (iv) involves TrkA-dependent mechanisms because the effects of NGF reapplication are blocked by acute exposure to specific and cell-permeable inhibitor of its high-affinity receptor. Taken together, this study may have important clinical implications in the field of AD neurodegeneration because it: (i) provides new insights on the earliest molecular mechanisms underlying the loss of synaptic/trafficking proteins and, then, of synapes integrity which occurs in vulnerable basal forebrain population at preclinical stages of neuropathology; (ii) offers prime presynaptic-based molecular target to extend the therapeutic time-window of NGF action in the strategy of improving its neuroprotective in vivo intervention in affected patients.
Collapse
Affiliation(s)
- Valentina Latina
- Institute of Translational Pharmacology, National Research Council (CNR) Rome, Italy
| | | | - Cristina Zona
- IRCCS Santa Lucia FoundationRome, Italy; Department of Systems Medicine, University of Rome Tor VergataRome, Italy
| | - Maria T Ciotti
- NGF and Molecular Mechanisms of Neurodegenerative Diseases, European Brain Research Institute (EBRI) Rome, Italy
| | - Giuseppina Amadoro
- Institute of Translational Pharmacology, National Research Council (CNR)Rome, Italy; NGF and Molecular Mechanisms of Neurodegenerative Diseases, European Brain Research Institute (EBRI)Rome, Italy
| | - Pietro Calissano
- NGF and Molecular Mechanisms of Neurodegenerative Diseases, European Brain Research Institute (EBRI) Rome, Italy
| |
Collapse
|
43
|
Canu N, Pagano I, La Rosa LR, Pellegrino M, Ciotti MT, Mercanti D, Moretti F, Sposato V, Triaca V, Petrella C, Maruyama IN, Levi A, Calissano P. Association of TrkA and APP Is Promoted by NGF and Reduced by Cell Death-Promoting Agents. Front Mol Neurosci 2017; 10:15. [PMID: 28197073 PMCID: PMC5281621 DOI: 10.3389/fnmol.2017.00015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 01/11/2017] [Indexed: 12/31/2022] Open
Abstract
The amyloid precursor protein (APP) interacts with the tropomyosin receptor kinase A (TrkA) in normal rat, mouse, and human brain tissue but not in Alzheimer's disease (AD) brain tissue. However, it has not been reported whether the two proteins interact directly, and if so, which domains are involved. Clarifying these points will increase our understanding of the role and regulation of the TrkA/APP interaction in normal brain functioning as well as in AD. Here we addressed these questions using bimolecular fluorescence complementation (BiFC) and the proximity ligation assay (PLA). We demonstrated that exogenously expressed APP and TrkA associate through their juxtamembrane/transmembrane domains, to form a complex that localizes mainly to the plasma membrane, endoplasmic reticulum (ER) and Golgi. Formation of the complex was inhibited by p75NTR, ShcC and Mint-2. Importantly, we demonstrated that the association between endogenous APP and TrkA in primary septal neurons were modified by NGF, or by drugs that either inhibit ER-to-Golgi transport or perturb microtubules and microfilaments. Interestingly, several agents that induce cell death [amyloid β (Aβ)-peptide, staurosporine and rapamycin], albeit via different mechanisms, all caused dissociation of APP/TrkA complexes and increased production of C-terminal fragment (β-CTF) APP fragment. These findings open new perspectives for investigating the interplay between these proteins during neurodegeneration and AD.
Collapse
Affiliation(s)
- Nadia Canu
- Department of System Medicine, University of Rome "Tor Vergata"Rome, Italy; Institute of Cellular Biology and Neurobiology, National Council of Research of RomeRome, Italy
| | - Ilaria Pagano
- Institute of Cellular Biology and Neurobiology, National Council of Research of Rome Rome, Italy
| | - Luca Rosario La Rosa
- Institute of Cellular Biology and Neurobiology, National Council of Research of Rome Rome, Italy
| | - Marsha Pellegrino
- Institute of Cellular Biology and Neurobiology, National Council of Research of Rome Rome, Italy
| | - Maria Teresa Ciotti
- Institute of Cellular Biology and Neurobiology, National Council of Research of Rome Rome, Italy
| | - Delio Mercanti
- Institute of Cellular Biology and Neurobiology, National Council of Research of Rome Rome, Italy
| | - Fabiola Moretti
- Institute of Cellular Biology and Neurobiology, National Council of Research of Rome Rome, Italy
| | - Valentina Sposato
- Institute of Cellular Biology and Neurobiology, National Council of Research of RomeRome, Italy; European Brain Research InstituteRome, Italy
| | - Viviana Triaca
- Institute of Cellular Biology and Neurobiology, National Council of Research of RomeRome, Italy; European Brain Research InstituteRome, Italy
| | - Carla Petrella
- Institute of Cellular Biology and Neurobiology, National Council of Research of Rome Rome, Italy
| | - Ichiro N Maruyama
- Information Processing Biology Unit, Okinawa Institute of Science and Technology Graduate University Okinawa, Japan
| | - Andrea Levi
- Institute of Cellular Biology and Neurobiology, National Council of Research of Rome Rome, Italy
| | - Pietro Calissano
- European Brain Research InstituteRome, Italy; Institute of Cellular Biology and Neurobiology, National Council of Research of RomeRome, Italy
| |
Collapse
|
44
|
Lou G, Zhang Q, Xiao F, Xiang Q, Su Z, Huang Y. Intranasal TAT-haFGF Improves Cognition and Amyloid-β Pathology in an AβPP/PS1 Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2016; 51:985-90. [PMID: 26890786 DOI: 10.3233/jad-151121] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Neurotoxic amyloid-β (Aβ) peptide causing cognitive function disabilities is one of the most characteristic pathological features in Alzheimer's disease (AD). A novel fusion protein, TAT-haFGF, was administrated to AβPP/PS1 transgenic mice by intravenous (IV) injection and intranasal (IN) delivery, respectively, for 5 weeks to compare the pharmacodynamics between the two routes of administration. Our results showed that IN administration of TAT-haFGF improved cognition and reduced Aβ plaques more significantly in AβPP/PS1 mice, when compared with IV injection. Our new findings suggest that TAT-haFGF might be a promising new therapy to attenuate AD pathological process.
Collapse
Affiliation(s)
- Guofeng Lou
- Institute of Biomedicine & Department of Cell Biology, Jinan University, Guangzhou, China
| | - Qihao Zhang
- Institute of Biomedicine & Department of Cell Biology, Jinan University, Guangzhou, China
| | - Fei Xiao
- Department of Pharmacology, Jinan University, Guangzhou, China
| | - Qi Xiang
- Institute of Biomedicine & Department of Cell Biology, Jinan University, Guangzhou, China
| | - Zhijian Su
- Institute of Biomedicine & Department of Cell Biology, Jinan University, Guangzhou, China
| | - Yadong Huang
- Institute of Biomedicine & Department of Cell Biology, Jinan University, Guangzhou, China
| |
Collapse
|
45
|
Intranasal Delivery of a Caspase-1 Inhibitor in the Treatment of Global Cerebral Ischemia. Mol Neurobiol 2016; 54:4936-4952. [PMID: 27520275 DOI: 10.1007/s12035-016-0034-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 08/01/2016] [Indexed: 02/07/2023]
Abstract
Caspase-1 is an enzyme implicated in neuroinflammation, a critical component of many diseases that affect neuronal degeneration. However, it is unknown whether a caspase-1 inhibitor can modify apoptotic neuronal damage incurred during transient global cerebral ischemia (GCI) and whether intranasal administration of a caspase-1 inhibitor is an effective treatment following GCI. The present study was conducted to examine the potential efficiency of post-ischemic intranasal administration of the caspase-1 inhibitor Boc-D-CMK in a 4-vessel occlusion model of GCI in the rat. Herein, we show that intranasal Boc-D-CMK readily penetrated the central nervous system, subsequently inhibiting caspase-1 activity, decreasing mitochondrial dysfunction, and attenuating caspase-3-dependent apoptotic pathway in ischemia-vulnerable hippocampal CA1 region. Further investigation regarding the mechanisms underlying Boc-D-CMK's neuroprotective effects revealed marked inhibition of reactive gliosis, as well as reduction of the neuroinflammatory response via inhibition of the downstream pro-inflammatory cytokine production. Intranasal Boc-D-CMK post-treatment also significantly enhanced the numbers of NeuN-positive cells while simultaneously decreasing the numbers of TUNEL-positive and PARP1-positive cells in hippocampal CA1. Correspondingly, behavioral tests showed that deteriorations in spatial learning and memory performance, and long-term recognition memory following GCI were significantly improved in the Boc-D-CMK post-treated animals. In summary, the current study demonstrates that the caspase-1 inhibitor Boc-D-CMK coordinates anti-inflammatory and anti-apoptotic actions to attenuate neuronal death in the hippocampal CA1 region following GCI. Furthermore, our data suggest that pharmacological inhibition of caspase-1 is a promising neuroprotective strategy to target ischemic neuronal injury and functional deficits following transient GCI.
Collapse
|
46
|
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]
|
47
|
Takito J, Kimura J, Kajima K, Uozumi N, Watanabe M, Yokosuka A, Mimaki Y, Nakamura M, Ohizumi Y. Nerve growth factor enhances the CRE-dependent transcriptional activity activated by nobiletin in PC12 cells. Can J Physiol Pharmacol 2016; 94:728-33. [PMID: 27128150 DOI: 10.1139/cjpp-2015-0394] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Prevention and treatment of Alzheimer disease are urgent problems for elderly people in developed countries. We previously reported that nobiletin, a poly-methoxylated flavone from the citrus peel, improved the symptoms in various types of animal models of memory loss and activated the cAMP responsive element (CRE)-dependent transcription in PC12 cells. Nobiletin activated the cAMP/PKA/MEK/Erk/MAPK signaling pathway without using the TrkA signaling activated by nerve growth factor (NGF). Here, we examined the effect of combination of nobiletin and NGF on the CRE-dependent transcription in PC12 cells. Although NGF alone had little effect on the CRE-dependent transcription, NGF markedly enhanced the CRE-dependent transcription induced by nobiletin. The NGF-induced enhancement was neutralized by a TrkA antagonist, K252a. This effect of NGF was effective on the early signaling event elicited by nobiletin. These results suggested that there was crosstalk between NGF and nobiletin signaling in activating the CRE-dependent transcription in PC12 cells.
Collapse
Affiliation(s)
- Jiro Takito
- a Department of Oral Anatomy and Developmental Biology, School of Dentistry, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
| | - Junko Kimura
- b Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Japan
| | - Koji Kajima
- c Sankyo Holdings Co., Ltd., 573-13 Denbou, Fuji-shi, Shizuoka, Japan
| | - Nobuyuki Uozumi
- d Department of Biomolecular Engineering, Graduate School of Engineering, Tohoku University, 6-6-07 Aobayama, Sendai, Miyagi, Japan
| | - Makoto Watanabe
- e Kansei Fukushi Research Center, Tohoku Fukushi University, 6-149-1 Kunimigaoka, Aoba-ku, Sendai, Miyagi, Japan
| | - Akihito Yokosuka
- f Laboratory of Medicinal Plant Science, School of Pharmacy, Tokyo University of Pharmacy and Life Science, 1432-1 Horinouchi, Hachioji, Tokyo, Japan
| | - Yoshihiro Mimaki
- f Laboratory of Medicinal Plant Science, School of Pharmacy, Tokyo University of Pharmacy and Life Science, 1432-1 Horinouchi, Hachioji, Tokyo, Japan
| | - Masanori Nakamura
- a Department of Oral Anatomy and Developmental Biology, School of Dentistry, Showa University, 1-5-8 Hatanodai, Shinagawa, Tokyo 142-8555, Japan
| | - Yasushi Ohizumi
- b Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, Japan.,e Kansei Fukushi Research Center, Tohoku Fukushi University, 6-149-1 Kunimigaoka, Aoba-ku, Sendai, Miyagi, Japan
| |
Collapse
|
48
|
Goodwin CR, Xu R, Iyer R, Sankey EW, Liu A, Abu-Bonsrah N, Sarabia-Estrada R, Frazier JL, Sciubba DM, Jallo GI. Local delivery methods of therapeutic agents in the treatment of diffuse intrinsic brainstem gliomas. Clin Neurol Neurosurg 2016; 142:120-127. [PMID: 26849840 DOI: 10.1016/j.clineuro.2016.01.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 01/05/2016] [Indexed: 10/22/2022]
Abstract
Brainstem gliomas comprise 10-20% of all pediatric central nervous system (CNS) tumors and diffuse intrinsic pontine gliomas (DIPGs) account for the majority of these lesions. DIPG is a rapidly progressive disease with almost universally fatal outcomes and a median survival less than 12 months. Current standard-of-care treatment for DIPG includes radiation therapy, but its long-term survival effects are still under debate. Clinical trials investigating the efficacy of systemic administration of various therapeutic agents have been associated with disappointing outcomes. Recent efforts have focused on improvements in chemotherapeutic agents employed and in methods of localized and targeted drug delivery. This review provides an update on current preclinical and clinical studies investigating treatment options for brainstem gliomas.
Collapse
Affiliation(s)
- C Rory Goodwin
- The Johns Hopkins University School of Medicine, Department of Neurosurgery, Baltimore, MD, USA
| | - Risheng Xu
- The Johns Hopkins University School of Medicine, Department of Neurosurgery, Baltimore, MD, USA
| | - Rajiv Iyer
- The Johns Hopkins University School of Medicine, Department of Neurosurgery, Baltimore, MD, USA
| | - Eric W Sankey
- The Johns Hopkins University School of Medicine, Department of Neurosurgery, Baltimore, MD, USA
| | - Ann Liu
- The Johns Hopkins University School of Medicine, Department of Neurosurgery, Baltimore, MD, USA
| | - Nancy Abu-Bonsrah
- The Johns Hopkins University School of Medicine, Department of Neurosurgery, Baltimore, MD, USA
| | - Rachel Sarabia-Estrada
- The Johns Hopkins University School of Medicine, Department of Neurosurgery, Baltimore, MD, USA
| | - James L Frazier
- The Johns Hopkins University School of Medicine, Department of Neurosurgery, Baltimore, MD, USA
| | - Daniel M Sciubba
- The Johns Hopkins University School of Medicine, Department of Neurosurgery, Baltimore, MD, USA
| | - George I Jallo
- The Johns Hopkins University School of Medicine, Department of Neurosurgery, Baltimore, MD, USA.
| |
Collapse
|
49
|
Lee JY, Park Y, Pun S, Lee SS, Lo JF, Lee LP. Real-time investigation of cytochrome c release profiles in living neuronal cells undergoing amyloid beta oligomer-induced apoptosis. NANOSCALE 2015; 7:10340-10343. [PMID: 26009283 DOI: 10.1039/c5nr02390d] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Intracellular Cyt c release profiles in living human neuroblastoma undergoing amyloid β oligomer (AβO)-induced apoptosis, as a model Alzheimer's disease-associated pathogenic molecule, were analysed in a real-time manner using plasmon resonance energy transfer (PRET)-based spectroscopy.
Collapse
Affiliation(s)
- Jae Young Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology, Gwangju 500-715, Republic of Korea
| | | | | | | | | | | |
Collapse
|
50
|
Nanoparticle-mediated growth factor delivery systems: A new way to treat Alzheimer's disease. J Control Release 2015; 206:187-205. [DOI: 10.1016/j.jconrel.2015.03.024] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 03/19/2015] [Accepted: 03/20/2015] [Indexed: 01/03/2023]
|