1
|
Keshri PK, Singh SP. Unraveling the AKT/ERK cascade and its role in Parkinson disease. Arch Toxicol 2024; 98:3169-3190. [PMID: 39136731 DOI: 10.1007/s00204-024-03829-9] [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: 04/02/2024] [Accepted: 07/25/2024] [Indexed: 09/17/2024]
Abstract
Parkinson disease represents a significant and growing burden on global healthcare systems, necessitating a deeper understanding of their underlying molecular mechanisms for the development of effective treatments. The AKT and ERK pathways play crucial roles in the disease, influencing multiple cellular pathways that support neuronal survival. Researchers have made notable progress in uncovering how these pathways are controlled by upstream kinases and how their downstream effects contribute to cell signalling. However, as we delve deeper into their intricacies, we encounter increasing complexity, compounded by the convergence of multiple signalling pathways. Many of their targets overlap with those of other kinases, and they not only affect specific substrates but also influence entire signalling networks. This review explores the intricate interplay of the AKT/ERK pathways with several other signalling cascades, including oxidative stress, endoplasmic reticulum stress, calcium homeostasis, inflammation, and autophagy, in the context of Parkinson disease. We discuss how dysregulation of these pathways contributes to disease progression and neuronal dysfunction, highlighting potential therapeutic targets for intervention. By elucidating the complex network of interactions between the AKT/ERK pathways and other signalling cascades, this review aims to provide insights into the pathogenesis of Parkinson disease and describe the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Priyanka Kumari Keshri
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India
| | - Surya Pratap Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, Uttar Pradesh, India.
| |
Collapse
|
2
|
Bernardes CP, Lopes Pinheiro E, Ferreira IG, de Oliveira IS, dos Santos NAG, Sampaio SV, Arantes EC, dos Santos AC. Fraction of C. d. collilineatus venom containing crotapotin protects PC12 cells against MPP + toxicity by activating the NGF-signaling pathway. J Venom Anim Toxins Incl Trop Dis 2024; 30:e20230056. [PMID: 38915449 PMCID: PMC11194915 DOI: 10.1590/1678-9199-jvatitd-2023-0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 05/08/2024] [Indexed: 06/26/2024] Open
Abstract
Background Parkinson's disease (PD) is the second most prevalent neurodegenerative disease. There is no effective treatment for neurodegenerative diseases. Snake venoms are a cocktail of proteins and peptides with great therapeutic potential and might be useful in the treatment of neurodegenerative diseases. Crotapotin is the acid chain of crotoxin, the major component of Crotalus durissus collilineatus venom. PD is characterized by low levels of neurotrophins, and synaptic and axonal degeneration; therefore, neurotrophic compounds might delay the progression of PD. The neurotrophic potential of crotapotin has not been studied yet. Methods We evaluated the neurotrophic potential of crotapotin in untreated PC12 cells, by assessing the induction of neurite outgrowth. The activation of the NGF signaling pathway was investigated through pharmacological inhibition of its main modulators. Additionally, its neuroprotective and neurorestorative effects were evaluated by assessing neurite outgrowth and cell viability in PC12 cells treated with the dopaminergic neurotoxin MPP+ (1-methyl-4-phenylpyridinium), known to induce Parkinsonism in humans and animal models. Results Crotapotin induced neuritogenesis in PC12 cells through the NGF-signaling pathway, more specifically, by activating the NGF-selective receptor trkA, and the PI3K/Akt and the MAPK/ERK cascades, which are involved in neuronal survival and differentiation. In addition, crotapotin had no cytotoxic effect and protected PC12 cells against the inhibitory effects of MPP+ on cell viability and differentiation. Conclusion These findings show, for the first time, that crotapotin has neurotrophic/neuroprotective/neurorestorative potential and might be beneficial in Parkinson's disease. Additional studies are necessary to evaluate the toxicity of crotapotin in other cell models.
Collapse
Affiliation(s)
- Carolina Petri Bernardes
- Department of Clinical Analyses, Toxicology and Food Science, School
of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP),
Ribeirão Preto, SP, Brazil
| | - Ernesto Lopes Pinheiro
- Department of Biomolecular Sciences, School of Pharmaceutical
Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP,
Brazil
| | - Isabela Gobbo Ferreira
- Department of Biomolecular Sciences, School of Pharmaceutical
Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP,
Brazil
| | - Isadora Sousa de Oliveira
- Department of Biomolecular Sciences, School of Pharmaceutical
Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP,
Brazil
| | - Neife Aparecida Guinaim dos Santos
- Department of Clinical Analyses, Toxicology and Food Science, School
of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP),
Ribeirão Preto, SP, Brazil
| | - Suely Vilela Sampaio
- Department of Clinical Analyses, Toxicology and Food Science, School
of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP),
Ribeirão Preto, SP, Brazil
| | - Eliane Candiani Arantes
- Department of Biomolecular Sciences, School of Pharmaceutical
Sciences of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, SP,
Brazil
| | - Antonio Cardozo dos Santos
- Department of Clinical Analyses, Toxicology and Food Science, School
of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (USP),
Ribeirão Preto, SP, Brazil
| |
Collapse
|
3
|
Yao C, Zhang H, Wang L, Li J. Correlation of serum Meteorin-like (Metrnl) level with type 2 diabetic peripheral neuropathy. BMC Endocr Disord 2024; 24:83. [PMID: 38849768 PMCID: PMC11162054 DOI: 10.1186/s12902-024-01616-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024] Open
Abstract
OBJECTIVE Meteorin-like (Metrnl), a secreted myokine, is a newly discovered neurotrophic factor. The aim of this study was to determine if there is a correlation between the Metrnl level and diabetic peripheral neuropathy (DPN). METHODS The investigation was conducted on a sample of 80 patients with type 2 diabetes mellitus (T2DM) and 60 healthy controls. The T2DM patients were categorized into two subgroups based on skin biopsy: the DPN subgroup (n = 20) and the diabetes without neuropathy subgroup (n = 60). RESULTS The T2DM groups had higher serum Metrnl concentrations compared with the controls. The serum Metrnl concentration was significantly lower in the DPN group than in T2DM patients without neuropathy. Logistic regression analysis demonstrated a notable correlation between serum Metrnl and DPN (OR: 0.997, 95% CI: 0.995-1.000, P < 0.05). Serum Metrnl level was negatively correlated with age and SBP after a simple logistic regression analysis. CONCLUSION Serum Metrnl concentration is independently correlated with DPN.
Collapse
Affiliation(s)
- Caixia Yao
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Endocrinology and Metabolism, Gaochun Hospital Affiliated to Jiangsu University, Nanjing, Jiangsu, China
| | - Hongman Zhang
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Endocrinology, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu, China
| | - Li Wang
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jianbo Li
- Department of Endocrinology and Metabolism, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
4
|
Patra P, Rani A, Sharma N, Mukherjee C, Jha HC. Unraveling the Connection of Epstein-Barr Virus and Its Glycoprotein M 146-157 Peptide with Neurological Ailments. ACS Chem Neurosci 2023. [PMID: 37290090 DOI: 10.1021/acschemneuro.3c00231] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023] Open
Abstract
Epstein-Barr virus (EBV) is known to be associated with several cancers along with neurological modalities like Alzheimer's disease (AD) and multiple sclerosis (MS). Previous study from our group revealed that a 12 amino acid peptide fragment (146SYKHVFLSAFVY157) of EBV glycoprotein M (gM) exhibits amyloid-like self-aggregative properties. In the current study, we have investigated its effect on Aβ42 aggregation along with its effect on neural cell immunology and disease markers. EBV virion was also considered for the above-mentioned investigation. An increase in the aggregation of Aβ42 peptide was observed upon incubation with gM146-157. Further, the exposure of EBV and gM146-157 onto neuronal cells indicated the upregulation of inflammatory molecules like IL-1β, IL-6, TNF-α, and TGF-β that suggested neuroinflammation. Besides, host cell factors like mitochondrial potential and calcium ion signaling play a crucial role in cellular homeostasis and alterations in these factors aid in neurodegeneration. Changes in mitochondrial membrane potential manifested a decrease while elevation in the level of total Ca2+ ions was observed. Amelioration of Ca2+ ions triggers excitotoxicity in neurons. Subsequently, neurological disease-associated genes APP, ApoE4, and MBP were found to be increased at the protein level. Additionally, demyelination of neurons is a hallmark of MS and the myelin sheath consists of ∼70% of lipid/cholesterol-associated moieties. Hereby, genes associated with cholesterol metabolism indicated changes at the mRNA level. Enhanced expression of neurotropic factors like NGF and BDNF was discerned postexposure to EBV and gM146-157. Altogether, this study delineates a direct connection of EBV and its peptide gM146-157 with neurological illnesses.
Collapse
Affiliation(s)
- Priyanka Patra
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore 453552, Madhya Pradesh, India
| | - Annu Rani
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore 453552, Madhya Pradesh, India
| | - Neha Sharma
- Department of Atomic Energy, Optical Coatings Laboratory, High Energy Lasers & Optics Section, Laser Technology Division, Laser Group, Raja Ramanna Centre for Advanced Technology, Indore 452013, Madhya Pradesh, India
| | - Chandrachur Mukherjee
- Department of Atomic Energy, Optical Coatings Laboratory, High Energy Lasers & Optics Section, Laser Technology Division, Laser Group, Raja Ramanna Centre for Advanced Technology, Indore 452013, Madhya Pradesh, India
| | - Hem Chandra Jha
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore 453552, Madhya Pradesh, India
| |
Collapse
|
5
|
Motaghinejad M, Gholami M, Emanuele E. Constant romantic feelings and experiences can protect against neurodegeneration: Potential role of oxytocin-induced nerve growth factor/protein kinase B/Cyclic response element-binding protein and nerve growth factor/protein kinase B/Phospholipase C-Gamma signaling pathways. BIOMEDICAL AND BIOTECHNOLOGY RESEARCH JOURNAL (BBRJ) 2023. [DOI: 10.4103/bbrj.bbrj_28_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
|
6
|
Expression and Signaling Pathways of Nerve Growth Factor (NGF) and Pro-NGF in Breast Cancer: A Systematic Review. Curr Oncol 2022; 29:8103-8120. [PMID: 36354700 PMCID: PMC9689427 DOI: 10.3390/curroncol29110640] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/19/2022] [Accepted: 10/25/2022] [Indexed: 11/07/2022] Open
Abstract
Breast cancer represents the most common type of cancer and is the leading cause of death due to cancer among women. Thus, the prevention and early diagnosis of breast cancer is of primary urgency, as well as the development of new treatments able to improve its prognosis. Nerve Growth Factor (NGF) is a neurotrophic factor involved in the regulation of neuronal functions through the binding of the Tropomyosin receptor kinase A (TrkA) and the Nerve Growth Factor receptor or Pan-Neurotrophin Receptor 75 (NGFR/p75NTR). In addition, its precursor (pro-NGF) can extert biological activity by forming a trimeric complex with NGFR/p75NTR and sortilin, or by binding to TrkA receptors with low affinity. Several examples of in vitro and in vivo evidence show that NGF is both synthesized and released by breast cancer cells, and has mitogen, antiapoptotic and angiogenic effects on these cells through the activation of different signaling cascades that involve TrkA and NGFR/p75NTR receptors. Conversely, pro-NGF signaling has been related to breast cancer invasion and metastasis. Other studies suggested that NGF and its receptors could represent a good diagnostic and prognostic tool, as well as promising therapeutic targets for breast cancer. In this paper, we comprehensively summarize and systematically review the current experimental evidence on this topic. INPLASY ID: INPLASY2022100017.
Collapse
|
7
|
Mattern L, Otten K, Miskey C, Fuest M, Izsvák Z, Ivics Z, Walter P, Thumann G, Johnen S. Molecular and Functional Characterization of BDNF-Overexpressing Human Retinal Pigment Epithelial Cells Established by Sleeping Beauty Transposon-Mediated Gene Transfer. Int J Mol Sci 2022; 23:12982. [PMID: 36361771 PMCID: PMC9656812 DOI: 10.3390/ijms232112982] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/30/2022] [Accepted: 10/25/2022] [Indexed: 04/12/2024] Open
Abstract
More and more patients suffer from multifactorial neurodegenerative diseases, such as age-related macular degeneration (AMD). However, their pathological mechanisms are still poorly understood, which complicates the development of effective therapies. To improve treatment of multifactorial diseases, cell-based gene therapy can be used to increase the expression of therapeutic factors. To date, there is no approved therapy for dry AMD, including late-stage geographic atrophy. We present a treatment option for dry AMD that transfers the brain-derived neurotrophic factor (BDNF) gene into retinal pigment epithelial (RPE) cells by electroporation using the plasmid-based Sleeping Beauty (SB) transposon system. ARPE-19 cells and primary human RPE cells were co-transfected with two plasmids encoding the SB100X transposase and the transposon carrying a BDNF transcription cassette. We demonstrated efficient expression and secretion of BDNF in both RPE cell types, which were further increased in ARPE-19 cell cultures exposed to hydrogen peroxide. BDNF-transfected cells exhibited lower apoptosis rates and stimulated neurite outgrowth in human SH-SY5Y cells. This study is an important step in the development of a cell-based BDNF gene therapy that could be applied as an advanced therapy medicinal product to treat dry AMD or other degenerative retinal diseases.
Collapse
Affiliation(s)
- Larissa Mattern
- Department of Ophthalmology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Katrin Otten
- Department of Ophthalmology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Csaba Miskey
- Division of Medical Biotechnology, Paul-Ehrlich-Institute, 63225 Langen, Germany
| | - Matthias Fuest
- Department of Ophthalmology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Zsuzsanna Izsvák
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Zoltán Ivics
- Division of Medical Biotechnology, Paul-Ehrlich-Institute, 63225 Langen, Germany
| | - Peter Walter
- Department of Ophthalmology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| | - Gabriele Thumann
- Department of Ophthalmology, University Hospitals of Geneva, 1205 Geneva, Switzerland
- Experimental Ophthalmology, University of Geneva, 1205 Geneva, Switzerland
| | - Sandra Johnen
- Department of Ophthalmology, University Hospital RWTH Aachen, 52074 Aachen, Germany
| |
Collapse
|
8
|
Padmakumar S, Kulkarni P, Ferris CF, Bleier BS, Amiji MM. Traumatic brain injury and the development of parkinsonism: Understanding pathophysiology, animal models, and therapeutic targets. Biomed Pharmacother 2022; 149:112812. [PMID: 35290887 PMCID: PMC9050934 DOI: 10.1016/j.biopha.2022.112812] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/06/2022] [Accepted: 03/08/2022] [Indexed: 02/06/2023] Open
Abstract
The clinical translation of therapeutic approaches to combat debilitating neurodegenerative conditions, such as Parkinson's disease (PD), remains as an urgent unmet challenge. The strong molecular association between the pathogenesis of traumatic brain injury (TBI) and the development of parkinsonism in humans has been well established. Therefore, a lot of ongoing research aims to investigate this pathology overlap in-depth, to exploit the common targets of TBI and PD for development of more effective and long-term treatment strategies. This review article intends to provide a detailed background on TBI pathophysiology and its established overlap with PD with an additional emphasis on the recent findings about their effect on perivascular clearance. Although, the traditional animal models of TBI and PD are still being considered, there is a huge focus on the development of combinatory hybrid animal models coupling concussion with the pre-established PD models for a better recapitulation of the human context of PD pathogenesis. Lastly, the therapeutic targets for TBI and PD, and the contemporary research involving exosomes, DNA vaccines, miRNA, gene therapy and gene editing for the development of potential candidates are discussed, along with the recent development of lesser invasive and promising central nervous system (CNS) drug delivery strategies.
Collapse
Affiliation(s)
- Smrithi Padmakumar
- Department of Pharmaceutical Sciences, School of Pharmacy and Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA, United States of America
| | - Praveen Kulkarni
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States of America
| | - Craig F Ferris
- Center for Translational NeuroImaging, Northeastern University, Boston, MA, United States of America
| | - Benjamin S Bleier
- Massachusetts Eye and Ear Infirmary, Harvard Medical School, Boston, MA, United States of America
| | - Mansoor M Amiji
- Department of Pharmaceutical Sciences, School of Pharmacy and Department of Chemical Engineering, College of Engineering, Northeastern University, Boston, MA, United States of America.
| |
Collapse
|
9
|
Bahlakeh G, Rahbarghazi R, Mohammadnejad D, Abedelahi A, Karimipour M. Current knowledge and challenges associated with targeted delivery of neurotrophic factors into the central nervous system: focus on available approaches. Cell Biosci 2021; 11:181. [PMID: 34641969 PMCID: PMC8507154 DOI: 10.1186/s13578-021-00694-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/28/2021] [Indexed: 12/23/2022] Open
Abstract
During the last decades, numerous basic and clinical studies have been conducted to assess the delivery efficiency of therapeutic agents into the brain and spinal cord parenchyma using several administration routes. Among conventional and in-progress administrative routes, the eligibility of stem cells, viral vectors, and biomaterial systems have been shown in the delivery of NTFs. Despite these manifold advances, the close association between the delivery system and regeneration outcome remains unclear. Herein, we aimed to discuss recent progress in the delivery of these factors and the pros and cons related to each modality.
Collapse
Affiliation(s)
- Gozal Bahlakeh
- Neurosciences Research Center (NSRC), Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Daruosh Mohammadnejad
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Abedelahi
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Karimipour
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran. .,Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
10
|
Stanga S, Boido M, Kienlen-Campard P. How to Build and to Protect the Neuromuscular Junction: The Role of the Glial Cell Line-Derived Neurotrophic Factor. Int J Mol Sci 2020; 22:ijms22010136. [PMID: 33374485 PMCID: PMC7794999 DOI: 10.3390/ijms22010136] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/07/2020] [Accepted: 12/16/2020] [Indexed: 12/12/2022] Open
Abstract
The neuromuscular junction (NMJ) is at the crossroad between the nervous system (NS) and the muscle. Following neurotransmitter release from the motor neurons (MNs), muscle contraction occurs and movement is generated. Besides eliciting muscle contraction, the NMJ represents a site of chemical bidirectional interplay between nerve and muscle with the active participation of Schwann cells. Indeed, signals originating from the muscle play an important role in synapse formation, stabilization, maintenance and function, both in development and adulthood. We focus here on the contribution of the Glial cell line-Derived Neurotrophic Factor (GDNF) to these processes and to its potential role in the protection of the NMJ during neurodegeneration. Historically related to the maintenance and survival of dopaminergic neurons of the substantia nigra, GDNF also plays a fundamental role in the peripheral NS (PNS). At this level, it promotes muscle trophism and it participates to the functionality of synapses. Moreover, compared to the other neurotrophic factors, GDNF shows unique peculiarities, which make its contribution essential in neurodegenerative disorders. While describing the known structural and functional changes occurring at the NMJ during neurodegeneration, we highlight the role of GDNF in the NMJ–muscle cross-talk and we review its therapeutic potential in counteracting the degenerative process occurring in the PNS in progressive and severe diseases such as Alzheimer’s disease (AD), Amyotrophic Lateral Sclerosis (ALS) and Spinal Muscular Atrophy (SMA). We also describe functional 3D neuromuscular co-culture systems that have been recently developed as a model for studying both NMJ formation in vitro and its involvement in neuromuscular disorders.
Collapse
Affiliation(s)
- Serena Stanga
- Department of Neuroscience Rita Levi Montalcini, University of Turin, 10126 Turin, Italy;
- Laboratory of Brain Development and Disease, Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano, Italy
- National Institute of Neuroscience (INN), 10125 Turin, Italy
- Correspondence:
| | - Marina Boido
- Department of Neuroscience Rita Levi Montalcini, University of Turin, 10126 Turin, Italy;
- Laboratory of Brain Development and Disease, Neuroscience Institute Cavalieri Ottolenghi, University of Turin, 10043 Orbassano, Italy
- National Institute of Neuroscience (INN), 10125 Turin, Italy
| | - Pascal Kienlen-Campard
- Institute of Neuroscience (IoNS), Université Catholique de Louvain (UCLouvain), 1200 Bruxelles, Belgium;
| |
Collapse
|
11
|
Faust K, Vajkoczy P, Xi B, Harnack D. The Effects of Deep Brain Stimulation of the Subthalamic Nucleus on Vascular Endothelial Growth Factor, Brain-Derived Neurotrophic Factor, and Glial Cell Line-Derived Neurotrophic Factor in a Rat Model of Parkinson's Disease. Stereotact Funct Neurosurg 2020; 99:256-266. [PMID: 33152730 DOI: 10.1159/000511121] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/23/2020] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Deep brain stimulation (DBS) of the subthalamic nucleus (STN) has evolved as a powerful therapeutic alternative for the treatment of Parkinson's disease (PD). Despite its clinical efficacy, the mechanisms of action have remained poorly understood. In addition to the immediate symptomatic effects, long-term neuroprotective effects have been suggested. Those may be mediated through neurotrophic factors (NFs) like vascular endothelial growth factor (VEGF), brain-derived neurotrophic factor (BDNF), and glial cell line-derived neurotrophic factor (GDNF). Here, the impact of DBS on the expression of NFs was analysed in a rat model of PD. METHODS Unilateral 6-hydroxydopamine (6-OHDA) lesioned rats received DBS in the STN using an implantable microstimulation system, sham DBS in the STN, or no electrode placement. Continuous unilateral STN-DBS (current intensity 50 µA, frequency 130 Hz, and pulse width 52 µs) was conducted for 14 days. Rats were then sacrificed and brains shock frozen. Striata and motor cortices were dissected with a cryostat. Levels of VEGF, BDNF, and GDNF were analysed, both by quantitative PCR and colorimetric ELISA. RESULTS PCR revealed a significant upregulation of only BDNF mRNA in the ipsilateral striata of the DBS group, when compared to the sham-stimulated group. There was no significant increase in VEGF mRNA or GDNF mRNA. ELISA analysis showed augmentations of BDNF, VEGF, as well as GDNF protein in the ipsilateral striata after DBS compared to sham stimulation. In the motor cortex, significant increases after DBS were observed for BDNF only, not for the other 2 NFs. CONCLUSIONS The upregulation of trophic factors induced by STN-DBS may participate in its long-term therapeutic efficacy and potentially neuroprotective effects.
Collapse
Affiliation(s)
- Katharina Faust
- Department of Neurosurgery, Charité University Hospital, Berlin, Germany,
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité University Hospital, Berlin, Germany
| | - Bai Xi
- Department of Neurosurgery, Charité University Hospital, Berlin, Germany
| | - Daniel Harnack
- Beelitz Neurology, Rehabilitation Clinic, Berlin, Germany
| |
Collapse
|
12
|
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
|
13
|
Agrawal M, Saraf S, Saraf S, Dubey SK, Puri A, Patel RJ, Ajazuddin, Ravichandiran V, Murty US, Alexander A. Recent strategies and advances in the fabrication of nano lipid carriers and their application towards brain targeting. J Control Release 2020; 321:372-415. [PMID: 32061621 DOI: 10.1016/j.jconrel.2020.02.020] [Citation(s) in RCA: 117] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/10/2020] [Accepted: 02/10/2020] [Indexed: 12/20/2022]
Abstract
In last two decades, the lipid nanocarriers have been extensively investigated for their drug targeting efficiency towards the critical areas of the human body like CNS, cardiac region, tumor cells, etc. Owing to the flexibility and biocompatibility, the lipid-based nanocarriers, including nanoemulsion, liposomes, SLN, NLC etc. have gained much attention among various other nanocarrier systems for brain targeting of bioactives. Across different lipid nanocarriers, NLC remains to be the safest, stable, biocompatible and cost-effective drug carrier system with high encapsulation efficiency. Drug delivery to the brain always remains a challenging issue for scientists due to the complex structure and various barrier mechanisms surrounding the brain. The application of a suitable nanocarrier system and the use of any alternative route of drug administration like nose-to-brain drug delivery could overcome the hurdle and improves the therapeutic efficiency of CNS acting drugs thereof. NLC, a second-generation lipid nanocarrier, upsurges the drug permeation across the BBB due to its unique structural properties. The biocompatible lipid matrix and nano-size make it an ideal drug carrier for brain targeting. It offers many advantages over other drug carrier systems, including ease of manufacturing and scale-up to industrial level, higher drug targeting, high drug loading, control drug release, compatibility with a wide range of drug substances, non-toxic and non-irritant behavior. This review highlights recent progresses towards the development of NLC for brain targeting of bioactives with particular reference to its surface modifications, formulations aspects, pharmacokinetic behavior and efficacy towards the treatment of various neurological disorders like AD, PD, schizophrenia, epilepsy, brain cancer, CNS infection (viral and fungal), multiple sclerosis, cerebral ischemia, and cerebral malaria. This work describes in detail the role and application of NLC, along with its different fabrication techniques and associated limitations. Specific emphasis is given to compile a summary and graphical data on the area explored by scientists and researchers worldwide towards the treatment of neurological disorders with or without NLC. The article also highlights a brief insight into two prime approaches for brain targeting, including drug delivery across BBB and direct nose-to-brain drug delivery along with the current global status of specific neurological disorders.
Collapse
Affiliation(s)
- Mukta Agrawal
- Rungta College of Pharmaceutical Sciences and Research, Kohka-Kurud Road, Bhilai, Chhattisgarh 490024, India
| | - Swarnlata Saraf
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, Chhattisgarh 492010, India
| | - Shailendra Saraf
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, Chhattisgarh 492010, India
| | - Sunil Kumar Dubey
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS-PILANI), Pilani Campus, Rajasthan, India
| | - Anu Puri
- RNA Structure and Design Section, RNA Biology Laboratory (RBL), Center for Cancer Research, NCI-Frederick, NIH, Frederick, USA
| | - Ravish J Patel
- Ramanbhai Patel College of Pharmacy (RPCP), Charotar University of Sciences and Technology (CHARUSAT), Gujarat 388421, India
| | - Ajazuddin
- Rungta College of Pharmaceutical Sciences and Research, Kohka-Kurud Road, Bhilai, Chhattisgarh 490024, India
| | - V Ravichandiran
- National Institute of Pharmaceutical Education and Research (NIPER-Kolkata), Ministry of Chemicals & Fertilizers, Govt. of India, Chunilal Bhawan 168, Maniktala Main Road, Kolkata 700054, India
| | - Upadhyayula Suryanarayana Murty
- National Institute of Pharmaceutical Education and Research (NIPER-Guwahati), Department of Pharmaceuticals, Ministry of Chemicals & Fertilizers, Govt. of India, NH 37, NITS Mirza, Kamrup, 781125 Guwahati, Assam, India
| | - Amit Alexander
- National Institute of Pharmaceutical Education and Research (NIPER-Guwahati), Department of Pharmaceuticals, Ministry of Chemicals & Fertilizers, Govt. of India, NH 37, NITS Mirza, Kamrup, 781125 Guwahati, Assam, India.
| |
Collapse
|
14
|
Delivanoglou N, Boziki M, Theotokis P, Kesidou E, Touloumi O, Dafi N, Nousiopoulou E, Lagoudaki R, Grigoriadis N, Charalampopoulos I, Simeonidou C. Spatio-temporal expression profile of NGF and the two-receptor system, TrkA and p75NTR, in experimental autoimmune encephalomyelitis. J Neuroinflammation 2020; 17:41. [PMID: 31996225 PMCID: PMC6990493 DOI: 10.1186/s12974-020-1708-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 01/09/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Nerve growth factor (NGF) and its receptors, tropomyosin receptor kinase A (TrkA) and pan-neurotrophin receptor p75 (p75NTR), are known to play bidirectional roles between the immune and nervous system. There are only few studies with inconclusive results concerning the expression pattern and role of NGF, TrkA, and p75NTR (NGF system) under the neuroinflammatory conditions in multiple sclerosis (MS) and its mouse model, the experimental autoimmune encephalomyelitis (EAE). The aim of this study is to investigate the temporal expression in different cell types of NGF system in the central nervous system (CNS) during the EAE course. METHODS EAE was induced in C57BL/6 mice 6-8 weeks old. CNS tissue samples were collected on specific time points: day 10 (D10), days 20-22 (acute phase), and day 50 (chronic phase), compared to controls. Real-time PCR, Western Blot, histochemistry, and immunofluorescence were performed throughout the disease course for the detection of the spatio-temporal expression of the NGF system. RESULTS Our findings suggest that both NGF and its receptors, TrkA and p75NTR, are upregulated during acute and chronic phase of the EAE model in the inflammatory lesions in the spinal cord. NGF and its receptors were co-localized with NeuN+ cells, GAP-43+ axons, GFAP+ cells, Arginase1+ cells, and Mac3+ cells. Furthermore, TrkA and p75NTR were sparsely detected on CNPase+ cells within the inflammatory lesion. Of high importance is our observation that despite EAE being a T-mediated disease, only NGF and p75NTR were shown to be expressed by B lymphocytes (B220+ cells) and no expression on T lymphocytes was noticed. CONCLUSION Our results indicate that the components of the NGF system are subjected to differential regulation during the EAE disease course. The expression pattern of NGF, TrkA, and p75NTR is described in detail, suggesting possible functional roles in neuroprotection, neuroregeneration, and remyelination by direct and indirect effects on the components of the immune system.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/metabolism
- Brain/pathology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Gene Expression Regulation/genetics
- Immunohistochemistry
- Mice
- Mice, Inbred C57BL
- Nerve Growth Factor/biosynthesis
- Nerve Growth Factor/genetics
- Receptor, trkA/biosynthesis
- Receptor, trkA/genetics
- Receptors, Nerve Growth Factor/biosynthesis
- Receptors, Nerve Growth Factor/genetics
- Spinal Cord/metabolism
- Spinal Cord/pathology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Nickoleta Delivanoglou
- Laboratory of Experimental Neurology and Neuroimmunology, B' Department of Neurology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Laboratory of Experimental Physiology, Department of Physiology and Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology, B' Department of Neurology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, B' Department of Neurology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology, B' Department of Neurology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
- Laboratory of Experimental Physiology, Department of Physiology and Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Olga Touloumi
- Laboratory of Experimental Neurology and Neuroimmunology, B' Department of Neurology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolina Dafi
- Laboratory of Experimental Neurology and Neuroimmunology, B' Department of Neurology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Evangelia Nousiopoulou
- Laboratory of Experimental Neurology and Neuroimmunology, B' Department of Neurology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Roza Lagoudaki
- Laboratory of Experimental Neurology and Neuroimmunology, B' Department of Neurology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, B' Department of Neurology, AHEPA University Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioannis Charalampopoulos
- Laboratory of Pharmacology, Department of Basic Sciences, School of Medicine, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation of Research and Technology Hellas, Heraklion, Greece
| | - Constantina Simeonidou
- Laboratory of Experimental Physiology, Department of Physiology and Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| |
Collapse
|
15
|
Dhakal S, Kushairi N, Phan CW, Adhikari B, Sabaratnam V, Macreadie I. Dietary Polyphenols: A Multifactorial Strategy to Target Alzheimer's Disease. Int J Mol Sci 2019; 20:E5090. [PMID: 31615073 PMCID: PMC6834216 DOI: 10.3390/ijms20205090] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/11/2019] [Accepted: 10/11/2019] [Indexed: 02/06/2023] Open
Abstract
Ageing is an inevitable fundamental process for people and is their greatest risk factor for neurodegenerative disease. The ageing processes bring changes in cells that can drive the organisms to experience loss of nutrient sensing, disrupted cellular functions, increased oxidative stress, loss of cellular homeostasis, genomic instability, accumulation of misfolded protein, impaired cellular defenses and telomere shortening. Perturbation of these vital cellular processes in neuronal cells can lead to life threatening neurological disorders like Alzheimer's Disease, Parkinson's Disease, Huntington's Disease, Lewy body dementia, etc. Alzheimer's Disease is the most frequent cause of deaths in the elderly population. Various therapeutic molecules have been designed to overcome the social, economic and health care burden caused by Alzheimer's Disease. Almost all the chemical compounds in clinical practice have been found to treat symptoms only limiting them to palliative care. The reason behind such imperfect drugs may result from the inefficiencies of the current drugs to target the cause of the disease. Here, we review the potential role of antioxidant polyphenolic compounds that could possibly be the most effective preventative strategy against Alzheimer's Disease.
Collapse
Affiliation(s)
- Sudip Dhakal
- School of Science, RMIT University, Bundoora, Victoria 3083, Australia.
| | - Naufal Kushairi
- Mushroom Research Centre, University of Malaya, 50603 Kuala Lumpur, Malaysia.
- Department of Anatomy, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Chia Wei Phan
- Mushroom Research Centre, University of Malaya, 50603 Kuala Lumpur, Malaysia.
- Department of Pharmaceutical Life Sciences, Faculty of Pharmacy, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Benu Adhikari
- School of Science, RMIT University, Bundoora, Victoria 3083, Australia.
| | - Vikineswary Sabaratnam
- Mushroom Research Centre, University of Malaya, 50603 Kuala Lumpur, Malaysia.
- Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Ian Macreadie
- School of Science, RMIT University, Bundoora, Victoria 3083, Australia.
| |
Collapse
|
16
|
FGF2 and dual agonist of NCAM and FGF receptor 1, Enreptin, rescue neurite outgrowth loss in hippocampal neurons expressing mutated huntingtin proteins. J Neural Transm (Vienna) 2019; 126:1493-1500. [PMID: 31501979 DOI: 10.1007/s00702-019-02073-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 08/30/2019] [Indexed: 10/26/2022]
Abstract
In the present study, we developed an in vitro model of Huntington disease (HD) by transfecting primary rat hippocampal neurons with plasmids coding for m-htt exon 1 with different number of CAG repeats (18, 50 and 115) and demonstrated the influence of the length of polyQ sequence on neurite elongation. We found that exogenously applied FGF2 significantly rescued the m-htt-induced loss of neurite outgrowth. Moreover, the Enreptin peptide, an FGFR1 and NCAM dual agonist, had a similar neuritogenic effect to FGF2 in clinically relevant m-htt 50Q-expressing neurons. This study has developed an in vitro model of primary hippocampal neurons transfected with m-htt-coding vectors that is a powerful tool to study m-htt-related effects on neuronal placticity.
Collapse
|
17
|
Claes M, De Groef L, Moons L. Target-Derived Neurotrophic Factor Deprivation Puts Retinal Ganglion Cells on Death Row: Cold Hard Evidence and Caveats. Int J Mol Sci 2019; 20:E4314. [PMID: 31484425 PMCID: PMC6747494 DOI: 10.3390/ijms20174314] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 08/28/2019] [Accepted: 08/30/2019] [Indexed: 12/14/2022] Open
Abstract
Glaucoma and other optic neuropathies are characterized by axonal transport deficits. Axonal cargo travels back and forth between the soma and the axon terminus, a mechanism ensuring homeostasis and the viability of a neuron. An example of vital molecules in the axonal cargo are neurotrophic factors (NTFs). Hindered retrograde transport can cause a scarcity of those factors in the retina, which in turn can tilt the fate of retinal ganglion cells (RGCs) towards apoptosis. This postulation is one of the most widely recognized theories to explain RGC death in the disease progression of glaucoma and is known as the NTF deprivation theory. For several decades, research has been focused on the use of NTFs as a novel neuroprotective glaucoma treatment. Until now, results in animal models have been promising, but translation to the clinic has been highly disappointing. Are we lacking important knowledge to lever NTF therapies towards the therapeutic armamentarium? Or did we get the wrong end of the stick regarding the NTF deprivation theory? In this review, we will tackle the existing evidence and caveats advocating for and against the target-derived NTF deprivation theory in glaucoma, whilst digging into associated therapy efforts.
Collapse
Affiliation(s)
- Marie Claes
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Lies De Groef
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium
| | - Lieve Moons
- Laboratory of Neural Circuit Development and Regeneration, Department of Biology, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
18
|
Izadpanah M, Seddigh A, Ebrahimi Barough S, Fazeli SAS, Ai J. Potential of Extracellular Vesicles in Neurodegenerative Diseases: Diagnostic and Therapeutic Indications. J Mol Neurosci 2018; 66:172-179. [PMID: 30140997 DOI: 10.1007/s12031-018-1135-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 07/20/2018] [Indexed: 01/09/2023]
Abstract
Extracellular vesicles (EVs) are membrane-bound vesicles, including exosomes and microvesicles. EVs are nanometer sized, found in physiological fluids such as urine, blood, cerebro-spinal fluid (CSF), with a capacity of transferring various biological materials such as microRNAs, proteins, and lipids among cells without direct cell-to-cell contact. Many cells in the nervous system have been shown to release EVs. These vesicles are involved in intercellular communication and a variety of biological processes such as modulation of immune response, signal transduction, and transport of genetic materials with low immunogenicity; therefore, they have also been recently investigated for the delivery of therapeutic molecules such as siRNAs and drugs in the treatment of diseases. In addition, since EV components reflect the physiological status of the cells and tissues producing them, they can be utilized as biomarkers for early detection of various diseases. In this review, we summarize EV application, in diagnosis as biomarker sources and as a carrier tool for drug delivery in EV-based therapies in neurodegenerative diseases.
Collapse
Affiliation(s)
- Mehrnaz Izadpanah
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, P. O. Box: 1417755469, Tehran, Iran.,Human and Animal Cell Bank, Iranian Biological Resource Center (IBRC), ACECR, Tehran, Iran
| | - Arshia Seddigh
- Department of Neurology, National Hospital for Neurology and Neurosurgery, University College London Hospitals NHS Trust, London, UK
| | - Somayeh Ebrahimi Barough
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, P. O. Box: 1417755469, Tehran, Iran
| | - Seyed Abolhassan Shahzadeh Fazeli
- Human and Animal Cell Bank, Iranian Biological Resource Center (IBRC), ACECR, Tehran, Iran.,Department of Molecular and Cellular Biology, Faculty of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, P. O. Box: 1417755469, Tehran, Iran.
| |
Collapse
|
19
|
Donsante A, Boulis NM. Progress in gene and cell therapies for the neuronal ceroid lipofuscinoses. Expert Opin Biol Ther 2018; 18:755-764. [PMID: 29936867 DOI: 10.1080/14712598.2018.1492544] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
INTRODUCTION The neuronal ceroid lipofuscinoses (NCLs) are a subset of lysosomal storage diseases (LSDs) that cause myoclonic epilepsy, loss of cognitive and motor function, degeneration of the retina leading to blindness, and early death. Most are caused by loss-of-function mutations in either lysosomal proteins or transmembrane proteins. Current therapies are supportive in nature. NCLs involving lysosomal enzymes are amenable to therapies that provide an exogenous source of protein, as has been used for other LSDs. Those that involve transmembrane proteins, however, require new approaches. AREAS COVERED This review will discuss potential gene and cell therapy approaches that have been, are, or may be in development for these disorders and those that have entered clinical trials. EXPERT OPINION In animal models, gene therapy approaches have produced remarkable improvements in neurological function and lifespan. However, a complete cure has not been reached for any NCL, and a better understanding of the limits of the current crop of vectors is needed to more fully address these diseases. The prospects for gene therapy, particularly those that can be delivered systemically and treat both the brain and peripheral tissue, are high. The future is beginning to look bright for NCL patients and their families.
Collapse
Affiliation(s)
- Anthony Donsante
- a Department of Neurosurgery , Emory University , Atlanta , GA , USA
| | - Nicholas M Boulis
- a Department of Neurosurgery , Emory University , Atlanta , GA , USA
| |
Collapse
|
20
|
Zang X, Cheng ZY, Sun Y, Hua N, Zhu LH, He L. The ameliorative effects and underlying mechanisms of dopamine D1-like receptor agonist SKF38393 on Aβ 1-42-induced cognitive impairment. Prog Neuropsychopharmacol Biol Psychiatry 2018; 81:250-261. [PMID: 28939187 DOI: 10.1016/j.pnpbp.2017.09.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/30/2017] [Accepted: 09/18/2017] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by extracellular amyloid plaques and intracellular neurofibrillary tangles. It is the most common form of human cognitive decline and dementia. In this study, we aim to systematically investigate the ameliorative effects of dopamine D1-like receptor agonist SKF38393 on cognitive dysfunction and explore its underlying mechanisms. The Aβ1-42 was injected intracerebroventricularly to establish cognitive disorder model. Then, a series of behavior tests were used. In order to further study the mechanisms, some relevant protein was assessed by ELISA method and Western blot. The results in behavior tests revealed that SKF38393 significantly ameliorated all the test indexes compared with the model mice. Then SKF38393 increased phosphorylation of cAMP response element binding protein (CREB) and expression of Bcl-2 in Western blot analyses. Furthermore, in ELISA assay, SKF38393 significantly increased the brain-derived neurotrophic factor (BDNF) levels and reduced the β-site APP cleaving enzyme1 (BACE1) and Aβ1-42 levels in hippocampus and cortex of mice. However, compared with SKF38393-H, all these results were significantly reversed by the dopamine D1 receptor antagonist SCH23390. These results indicated that SKF38393 could ameliorate Aβ1-42-induced cognitive dysfunction in mice, which may be related to D1 receptor activation. It leads to the phosphorylation of CREB, which promote the expression of BDNF, Bcl-2 and decrease the expression of Aβ1-42 of mice. Our findings suggest that dopamine D1-like receptor may be a potential target for the treatment of AD and its agonists may become a novel drug in the future.
Collapse
Affiliation(s)
- Xuan Zang
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Zhao-Yan Cheng
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Yi Sun
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Nan Hua
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Li-Hua Zhu
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
21
|
Yang EJ, Mahmood U, Kim H, Choi M, Choi Y, Lee JP, Chang MJ, Kim HS. Alterations in protein phosphorylation in the amygdala of the 5XFamilial Alzheimer's disease animal model. J Pharmacol Sci 2017; 133:261-267. [PMID: 28408165 DOI: 10.1016/j.jphs.2017.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 03/13/2017] [Accepted: 03/21/2017] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease is the most common disease underlying dementia in humans. Two major neuropathological hallmarks of AD are neuritic plaques primarily composed of amyloid beta peptide and neurofibrillary tangles primarily composed of hyperphosphorylated tau. In addition to impaired memory function, AD patients often display neuropsychiatric symptoms and abnormal emotional states such as confusion, delusion, manic/depressive episodes and altered fear status. Brains from AD patients show atrophy of the amygdala which is involved in fear expression and emotional processing as well as hippocampal atrophy. However, which molecular changes are responsible for the altered emotional states observed in AD remains to be elucidated. Here, we observed that the fear response as assessed by evaluating fear memory via a cued fear conditioning test was impaired in 5XFamilial AD (5XFAD) mice, an animal model of AD. Compared to wild-type mice, 5XFAD mice showed changes in the phosphorylation of twelve proteins in the amygdala. Thus, our study provides twelve potential protein targets in the amygdala that may be responsible for the impairment in fear memory in AD.
Collapse
Affiliation(s)
- Eun-Jeong Yang
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehakro, Jongro-gu, Seoul, Republic of Korea.
| | - Usman Mahmood
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehakro, Jongro-gu, Seoul, Republic of Korea.
| | - Hyunju Kim
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehakro, Jongro-gu, Seoul, Republic of Korea.
| | - Moonseok Choi
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehakro, Jongro-gu, Seoul, Republic of Korea.
| | - Yunjung Choi
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehakro, Jongro-gu, Seoul, Republic of Korea.
| | - Jean-Pyo Lee
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, 1430 Tulane Ave, SL99, New Orleans, LA 70112, USA.
| | - Moon-Jeong Chang
- Department of Foods and Nutrition, College of Natural Science, Kookmin University, Seoul, Republic of Korea.
| | - Hye-Sun Kim
- Department of Pharmacology and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehakro, Jongro-gu, Seoul, Republic of Korea; Seoul National University College of Medicine, Bundang Hospital, Bundang-Gu, Sungnam, Republic of Korea; Neuroscience Research Institute, College of Medicine, Seoul National University, 103 Daehakro, Jongro-gu, Seoul, Republic of Korea.
| |
Collapse
|
22
|
Mohibbullah M, Bhuiyan MMH, Hannan MA, Getachew P, Hong YK, Choi JS, Choi IS, Moon IS. The Edible Red Alga Porphyra yezoensis Promotes Neuronal Survival and Cytoarchitecture in Primary Hippocampal Neurons. Cell Mol Neurobiol 2016; 36:669-82. [PMID: 26259718 DOI: 10.1007/s10571-015-0247-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 07/28/2015] [Indexed: 12/13/2022]
Abstract
The edible red alga Porphyra yezoensis is among the most popular marine algae and is of economic and medicinal importance. In the present study, the neurotrophic and neuroprotective activities of the ethanol extract of P. yezoensis (PYE) were investigated in primary cultures of hippocampal neurons. Results revealed that PYE significantly increased neurite outgrowth at an optimal concentration of 15 µg/mL. PYE dose-dependently increased viable cells, significantly accelerated the rate of neuronal differentiation in cultures, promoted axodendritic arborization, and eventually induced synaptogenesis. In addition to morphological development, PYE also promoted functional maturation as indicated by the staining of live cultures with FM 1-43. Moreover, PYE increased neuronal survivability, which was attributed to reduced apoptosis and its ROS scavenging activity. Taurine, a major organic acid in PYE (2.584/100 mg of dry PYE) promoted neurite outgrowth in a dose-dependent manner, and this promotion was suppressed by the taurine antagonist isethionic acid. The study indicates that PYE and its active component, taurine, facilitate neuronal development and maturation and have a neuroprotective effect.
Collapse
Affiliation(s)
- Md Mohibbullah
- Department of Biotechnology, Pukyong National University, Namku, Busan, 608-737, Republic of Korea
| | | | - Md Abdul Hannan
- Department of Biotechnology, Pukyong National University, Namku, Busan, 608-737, Republic of Korea
- Department of Biochemistry and Molecular Biology, Bangladesh Agricultural University, Mymensingh, 2202, Bangladesh
| | - Paulos Getachew
- Department of Biotechnology, Pukyong National University, Namku, Busan, 608-737, Republic of Korea
| | - Yong-Ki Hong
- Department of Biotechnology, Pukyong National University, Namku, Busan, 608-737, Republic of Korea
| | - Jae-Suk Choi
- RIS Center, IACF, Silla University, Sasang-gu, Busan, 617-736, Republic of Korea
| | - In Soon Choi
- RIS Center, IACF, Silla University, Sasang-gu, Busan, 617-736, Republic of Korea
- Department of Biological Science, Silla University, Sasang-gu, Busan, 617-736, Republic of Korea
| | - Il Soo Moon
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju, 780-714, Republic of Korea.
| |
Collapse
|
23
|
Dunbar GL, Sandstrom MI, Rossignol J, Lescaudron L. Neurotrophic Enhancers as Therapy for Behavioral Deficits in Rodent Models of Huntington's Disease: Use of Gangliosides, Substituted Pyrimidines, and Mesenchymal Stem Cells. ACTA ACUST UNITED AC 2016; 5:63-79. [PMID: 16801683 DOI: 10.1177/1534582306289367] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The interest in using neurotrophic factors as potential treatments for neurodegenerative disorders, such as Huntington's disease, has grown in the past decade. A major impediment for the clinical utility of neurotrophic factors is their inability to cross the blood-brain barrier in therapeutically significant amounts. Although several novel mechanisms for delivering exogenous neurotrophins to the brain have been developed, most of them involve invasive procedures or present significant risks. One approach to circumventing these problems is using therapeutic agents that can be administered systemically and have the ability to enhance the activity of neurotrophic factors. This review highlights the use of gangliosides, substituted pyrimidines, and mesenchymal stem cells as neurotrophic enhancers that have significant therapeutic potential while avoiding the pitfalls of delivering exogenous neurotrophic factors through the blood-brain barrier. The review focuses on the potential of these neurotrophic enhancers for treating the behavioral deficits in rodent models of Huntington's disease.
Collapse
|
24
|
Hernando S, Gartziandia O, Herran E, Pedraz JL, Igartua M, Hernandez RM. Advances in nanomedicine for the treatment of Alzheimer’s and Parkinson’s diseases. Nanomedicine (Lond) 2016; 11:1267-85. [DOI: 10.2217/nnm-2016-0019] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Alzheimer‘s disease and Parkinson’s disease are the most common neurodegenerative diseases worldwide. Despite all the efforts made by the scientific community, current available treatments have limited effectiveness, without halting the progression of the disease. That is why, new molecules such as growth factors, antioxidants and metal chelators have been raised as new therapeutical approaches. However, these molecules have difficulties to cross the blood–brain barrier limiting its therapeutic effect. The development of nanometric drug delivery systems may permit a targeted and sustained release of old and new treatments offering a novel strategy to treat these neurodegenerative disorders. This review summarized the main investigated drug delivery systems as promising approaches to treat Alzheimer‘s disease and Parkinson’s disease.
Collapse
Affiliation(s)
- Sara Hernando
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01006, Spain
| | - Oihane Gartziandia
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01006, Spain
| | - Enara Herran
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01006, Spain
| | - Jose Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01006, Spain
| | - Manoli Igartua
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01006, Spain
| | - Rosa Maria Hernandez
- NanoBioCel Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Vitoria-Gasteiz 01006, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials & Nanomedicine (CIBER-BBN), Vitoria-Gasteiz 01006, Spain
| |
Collapse
|
25
|
Moosavi F, Hosseini R, Saso L, Firuzi O. Modulation of neurotrophic signaling pathways by polyphenols. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 10:23-42. [PMID: 26730179 PMCID: PMC4694682 DOI: 10.2147/dddt.s96936] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Polyphenols are an important class of phytochemicals, and several lines of evidence have demonstrated their beneficial effects in the context of a number of pathologies including neurodegenerative disorders such as Alzheimer’s and Parkinson’s disease. In this report, we review the studies on the effects of polyphenols on neuronal survival, growth, proliferation and differentiation, and the signaling pathways involved in these neurotrophic actions. Several polyphenols including flavonoids such as baicalein, daidzein, luteolin, and nobiletin as well as nonflavonoid polyphenols such as auraptene, carnosic acid, curcuminoids, and hydroxycinnamic acid derivatives including caffeic acid phentyl ester enhance neuronal survival and promote neurite outgrowth in vitro, a hallmark of neuronal differentiation. Assessment of underlying mechanisms, especially in PC12 neuronal-like cells, reveals that direct agonistic effect on tropomyosin receptor kinase (Trk) receptors, the main receptors of neurotrophic factors including nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) explains the action of few polyphenols such as 7,8-dihydroxyflavone. However, several other polyphenolic compounds activate extracellular signal-regulated kinase (ERK) and phosphoinositide 3-kinase (PI3K)/Akt pathways. Increased expression of neurotrophic factors in vitro and in vivo is the mechanism of neurotrophic action of flavonoids such as scutellarin, daidzein, genistein, and fisetin, while compounds like apigenin and ferulic acid increase cyclic adenosine monophosphate response element-binding protein (CREB) phosphorylation. Finally, the antioxidant activity of polyphenols reflected in the activation of Nrf2 pathway and the consequent upregulation of detoxification enzymes such as heme oxygenase-1 as well as the contribution of these effects to the neurotrophic activity have also been discussed. In conclusion, a better understanding of the neurotrophic effects of polyphenols and the concomitant modulations of signaling pathways is useful for designing more effective agents for management of neurodegenerative diseases.
Collapse
Affiliation(s)
- Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmacology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Razieh Hosseini
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Pharmacology, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Rome, Italy
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
26
|
XingNaoJing, prescription of traditional Chinese medicine, prevents autophagy in experimental stroke by repressing p53-DRAM pathway. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2015; 15:377. [PMID: 26481508 PMCID: PMC4617486 DOI: 10.1186/s12906-015-0882-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 09/28/2015] [Indexed: 12/17/2022]
Abstract
Background Xingnaojing (XNJ), a well known prescription in traditional Chinese medicine, has been used for treatment of stroke in China. However, the effects and mechanisms of XNJ on autophagy are not clear. Here, we used the cell models of autophagy induced by serum-free condition and ischemia stroke in rats to further investigate whether the p53-DRAM pathway is involved in the effects of XNJ on autophagy. Methods We used the cell model of autophagy induced by serum-free condition and the rat model of ischemia caused by a middle cerebral artery occlusion (MCAO). The effects of XNJ on p53 transcriptional activity of PC12 cells were evaluated by the luciferase activity assay. The mRNA levels and the expression of p53 and its target autophagy gene DRAM (damage-regulated autophagy modulator) were analyzed respectively by Quantitative-RTPCR and Western blot assay. The activation of autophagy was detected by the levels of autophagy markers, microtubule associated protein light chain 3 (LC3) and p62 by Immunofluorescence and Western blot. p53 inhibitor was used to determine whether p53 is responsible for the effects of XNJ on preventing autophagy. Results The assay for luciferase activity of p53 promoter indicated that XNJ inhibited p53 transcriptional activity. XNJ reduced the expression of p53 and its target autophagy gene DRAM (damage-regulated autophagy modulator) in serum-free condition PC12 cells and the cortex in MCAO rats. XNJ reduced autophagy of PC12 cells induced by serum-free condition and the cortex in MCAO rats. Furthermore, suppression of p53 by p53 inhibitor significantly reduced the effects of XNJ on the autophagy of PC12 cells in serum-free condition. Conclusion XNJ prevents autophagy in experimental stroke by repressing p53/DRAM pathway. Our findings are therefore of considerable therapeutic significance and provide the novel and potential application of XNJ for the treatment of brain diseases.
Collapse
|
27
|
Naito E, Kudo D, Sekine SI, Watanabe K, Kobatake Y, Tamaoki N, Inden M, Iida K, Ito Y, Hozumi I, Shibata T, Maeda S, Kamishina H. Characterization of canine dental pulp cells and their neuroregenerative potential. In Vitro Cell Dev Biol Anim 2015; 51:1012-22. [PMID: 26170225 DOI: 10.1007/s11626-015-9935-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 06/18/2015] [Indexed: 12/13/2022]
Abstract
Dental pulp cells (DPCs) of various species have been studied for their potentials of differentiation into functional neurons and secretion of neurotrophic factors. In canine, DPCs have only been studied for cell surface markers and differentiation, but there is little direct evidence for therapeutic potentials for neurological disorders. The present study aimed to further characterize canine DPCs (cDPCs), particularly focusing on their neuroregenerative potentials. It was also reported that superparamagnetic iron oxide (SPIO) particles were useful for labeling of MSCs and tracking with magnetic resonance imaging (MRI). Our data suggested that cDPCs hold higher proliferation capacity than bone marrow stromal cells, the other type of mesenchymal stem cells which have been the target of intensive research. Canine DPCs constitutively expressed neural markers, suggesting a close relationship to the nervous system in their developmental origin. Canine DPCs promoted neuritogenesis of PC12 cells, most likely through secretion of neurotrophic factors. Furthermore, SPIO nanoparticles could be effectively transported to cDPCs without significant cytotoxicity and unfavorable effects on neuritogenesis. SPIO-labeled cDPCs embedded in agarose spinal cord phantoms were successfully visualized with a magnetic resonance imaging arousing a hope for noninvasive cell tracking in transplantation studies.
Collapse
Affiliation(s)
- Eiji Naito
- Department of Veterinary Medicine, Faculty Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Daichi Kudo
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Shin-ichiro Sekine
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Kazuhiro Watanabe
- Department of Veterinary Medicine, Faculty Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Yui Kobatake
- Department of Veterinary Medicine, Faculty Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Naritaka Tamaoki
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Masatoshi Inden
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Kazuki Iida
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Yusuke Ito
- Department of Veterinary Medicine, Faculty Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Isao Hozumi
- Laboratory of Medical Therapeutics and Molecular Therapeutics, Gifu Pharmaceutical University, Gifu, Japan
| | - Toshiyuki Shibata
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Sadatoshi Maeda
- Department of Veterinary Medicine, Faculty Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan
| | - Hiroaki Kamishina
- Department of Veterinary Medicine, Faculty Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu, 501-1193, Japan.
| |
Collapse
|
28
|
Abstract
It is becoming increasingly clear that neurological diseases are multi-factorial involving disruptions in multiple cellular systems. Thus, while each disease has its own initiating mechanisms and pathologies, certain common pathways appear to be involved in most, if not all, neurological diseases. Thus, it is unlikely that modulating only a single factor will be effective at either preventing disease development or slowing disease progression. A better approach is to identify small (< 900 daltons) molecules that have multiple biological activities relevant to the maintenance of brain function. We have identified an orally active, novel neuroprotective and cognition-enhancing molecule, the flavonoid fisetin. Fisetin not only has direct antioxidant activity but it can also increase the intracellular levels of glutathione, the major intracellular antioxidant. Fisetin can also activate key neurotrophic factor signaling pathways. In addition, it has anti-inflammatory activity and inhibits the activity of lipoxygenases, thereby reducing the production of pro-inflammatory eicosanoids and their by-products. This wide range of actions suggests that fisetin has the ability to reduce the impact of age-related neurological diseases on brain function.
Collapse
Affiliation(s)
- Pamela Maher
- The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd, La Jolla, CA 92037,
| |
Collapse
|
29
|
dos Santos NAG, Martins NM, Silva RDB, Ferreira RS, Sisti FM, dos Santos AC. Caffeic acid phenethyl ester (CAPE) protects PC12 cells from MPP+ toxicity by inducing the expression of neuron-typical proteins. Neurotoxicology 2014; 45:131-8. [PMID: 25454720 DOI: 10.1016/j.neuro.2014.09.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 08/29/2014] [Accepted: 09/23/2014] [Indexed: 01/18/2023]
Abstract
Neurite loss is an early event in neurodegenerative diseases; therefore, the regeneration of the network of neurites constitutes an interesting strategy of treatment for such disorders. Neurotrophic factors play a critical role in neuronal regeneration, but their clinical use is limited by their inability to cross the blood brain barrier. Oxidative and inflammatory events are implicated in neurodegeneration and antioxidant compounds have been suggested as potential neuroprotectors. The protective potential of CAPE (caffeic acid phenethyl ester) has been shown in different models of neurotoxicity (in vitro and in vivo) and it has been associated with immune-modulatory, antioxidant and anti-inflammatory properties; however, other mechanisms might be involved. The present study demonstrates that CAPE protects PC12 cells from the cellular death induced by the dopaminergic neurotoxin MPP(+) by increasing the network of neurites. Results showed that CAPE induced the formation, elongation and ramification of neurites in PC12 cells non-stimulated with NGF (nerve growth factor) and inhibited the shortage of neurites induced by the dopaminergic neurotoxin. These effects were associated with increased expression of neuron-typical proteins responsible for axonal growth (GAP-43) and synaptogenesis (synaptophysin and synapsin I). It is noteworthy that, unlike neurotrophins, CAPE would be able to cross the blood brain barrier and exert its neurotrophic effects in the brain. This study corroborates the therapeutic potential of CAPE in neurodegenerative diseases while proposes the involvement of neuroplasticity in the mechanism of neuroprotection.
Collapse
Affiliation(s)
- Neife Aparecida Guinaim dos Santos
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil.
| | - Nádia Maria Martins
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Roberto de Barros Silva
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Rafaela Scalco Ferreira
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Flávia Malvestio Sisti
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Antonio Cardozo dos Santos
- Department of Clinical Analyses, Toxicology and Food Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
30
|
Shi H, Xie D, Yang R, Cheng Y. Synthesis of caffeic acid phenethyl ester derivatives, and their cytoprotective and neuritogenic activities in PC12 cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:5046-5053. [PMID: 24840770 DOI: 10.1021/jf500464k] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Twenty-one caffeic acid phenethyl ester (CAPE) derivatives were synthesized, and characterized by IR, HR-MS, (1)H and (13)C NMR analyses. All compounds were evaluated for their cytoprotective effects against H2O2-induced cytotoxicity and neuritogenic activities in the neurite outgrowth in PC12 cells. Compounds 1 and 20 exhibited stronger cytoprotective activities than their parent compound CAPE at 4 nM. Compounds 1, 4, 12 and 13 showed potential neuritogenic activities at 0.5 nM, while compounds 19 and 20 induced neurite outgrowth at 10 nM. The results from this study suggested that CAPE and its derivatives may be potential functional food ingredients for the prevention of neurodegenerative diseases.
Collapse
Affiliation(s)
- Haiming Shi
- Institute of Food and Nutraceutical Science, SJTU-Rich Research Institute of Nutrition and Skin Science, School of Agriculture and Biology, Shanghai Jiao Tong University , Shanghai 200240, China
| | | | | | | |
Collapse
|
31
|
Chen Y, Wei G, Nie H, Lin Y, Tian H, Liu Y, Yu X, Cheng S, Yan R, Wang Q, Liu DH, Deng W, Lai Y, Zhou JH, Zhang SX, Lin WW, Chen DF. β-Asarone prevents autophagy and synaptic loss by reducing ROCK expression in asenescence-accelerated prone 8 mice. Brain Res 2014; 1552:41-54. [DOI: 10.1016/j.brainres.2014.01.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 12/14/2013] [Accepted: 01/06/2014] [Indexed: 12/19/2022]
|
32
|
Chen JH, Lee DC, Chiu IM. Cytotoxic effects of acrylamide in nerve growth factor or fibroblast growth factor 1-induced neurite outgrowth in PC12 cells. Arch Toxicol 2013; 88:769-80. [PMID: 24318646 DOI: 10.1007/s00204-013-1174-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2013] [Accepted: 11/20/2013] [Indexed: 12/26/2022]
Abstract
Acrylamide is a neurological and reproductive toxicant in humans and laboratory animals; however, the neuron developmental toxicity of acrylamide remains unclear. The aims of this study are to investigate the cytotoxicity and neurite outgrowth inhibition of acrylamide in nerve growth factor (NGF)- or fibroblast growth factor 1 (FGF1)-mediated neural development of PC12 cells. MTS assay showed that acrylamide treatment suppresses NGF- or FGF1-induced PC12 cell proliferation in a time- and dose-dependent manner. Quantification of neurite outgrowth demonstrated that 0.5 mM acrylamide treatment resulted in significant decrease in differentiation of NGF- or FGF1-stimulated PC12 cells. This decrease is accompanied with the reduced expression of growth-associated protein-43, a neuronal marker. Moreover, relative levels of pERK, pAKT, pSTAT3 and pCREB were increased within 5-10 min when PC12 cells were treated with NGF or FGF1. Acrylamide (0.5 mM) decreases the NGF-induced activation of AKT-CREB but not ERK-STAT3 within 20 min. Similarly, acrylamide (0.5 mM) decreases the FGF1-induced activation of AKT-CREB within 20 min. In contrast to the NGF treatment, the ERK-STAT3 activation that was induced by FGF1 was slightly reduced by 0.5 mM acrylamide. We further showed that PI3K inhibitor (LY294002), but not MEK inhibitor (U0126), could synergize with acrylamide (0.5 mM) to reduce the cell viability and neurite outgrowth in NGF- or FGF1-stimulated PC12 cells. Moreover, acrylamide (0.5 mM) increased reactive oxygen species (ROS) activities in NGF- or FGF1-stimulated PC12 cells. This increase was reversed by Trolox (an ROS scavenging agent) co-treatment. Together, our findings reveal that NGF- or FGF1-stimulation of the neuronal differentiation of PC12 cells is attenuated by acrylamide through the inhibition of PI3K-AKT-CREB signaling, along with the production of ROS.
Collapse
Affiliation(s)
- Jong-Hang Chen
- Institute of Cellular and System Medicine, National Health Research Institutes, 35, Keyan Rd, Miaoli, 350, Taiwan
| | | | | |
Collapse
|
33
|
Leptin as a neuroprotective agent in glaucoma. Med Hypotheses 2013; 81:797-802. [PMID: 24035688 DOI: 10.1016/j.mehy.2013.08.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 08/05/2013] [Accepted: 08/22/2013] [Indexed: 11/22/2022]
Abstract
Glaucoma is a disease characterized by progressive optic nerve degeneration and is the leading cause of irreversible blindness worldwide. More than 60 million people globally are affected by glaucoma, of which 8 million people suffer from bilateral blindness, making glaucoma the second leading cause of bilateral blindness worldwide. Current management of glaucoma is aimed at reducing intraocular pressure via a number of different strategies. Current treatments do not attempt to correct the underlying pathology of glaucoma, which is the cell degeneration and ultimate death of retinal ganglion cells, thereby limiting their clinical efficacy. A neuroprotective approach to glaucoma management would address the underlying pathology and would, in theory, be beneficial to all patients regardless of risk and causative factors. Here it is proposed that leptin could be used as a potential neuroprotective agent in the management of glaucoma. Leptin has shown neuroprotective promise in a number of neurodegenerative diseases, and there has been increasing evidence that glaucomatous neurodegeneration is analogous to other neurodegenerative diseases in the central nervous system. Leptin could target retinal ganglion cell death by a number of mechanisms, namely apoptosis, oxidative stress and excitotoxicity reduction. This article presents evidence linking current understanding about leptin's neuroprotective effect and the molecular mechanisms underlying glaucoma.
Collapse
|
34
|
Lin LF, Liao MJ, Xue XY, Zhang W, Yan L, Cai L, Zhou XW, Zhou X, Luo HM. Combination of Aβ clearance and neurotrophic factors as a potential treatment for Alzheimer's disease. Neurosci Bull 2013; 29:111-20. [PMID: 23179066 PMCID: PMC5561854 DOI: 10.1007/s12264-012-1287-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 06/12/2012] [Indexed: 12/31/2022] Open
Abstract
There is no effective drug to treat Alzheimer's disease (AD), a neurodegenerative disease affecting an estimated 30 million people around the world. Strongly supported by preclinical and clinical studies, amyloid-beta (Aβ) may be a target for developing drugs against AD. Meanwhile, the fact that localized neuronal death/loss and synaptic impairment occur in AD should also be considered. Neuronal regeneration, which does not occur normally in the mammalian central nervous system, can be promoted by neurotrophic factors (NTFs). Evidence from clinical trials has shown that both Aβ clearance and NTFs are potentially effective in treating AD, thus a new approach combining Aβ clearance and administration of NTFs may be an effective therapeutic strategy.
Collapse
Affiliation(s)
- Lian-Feng Lin
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
| | - Min-Jing Liao
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
- Department of Laboratory Medicine, Medical College of Hunan Normal University, Changsha, 410013 China
| | - Xiao-Yan Xue
- Ganzhou People’s Hospital, Ganzhou, 341000 China
| | - Wei Zhang
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
| | - Li Yan
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
| | - Liang Cai
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
| | - Xiao-Wen Zhou
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
| | - Xing Zhou
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
| | - Huan-Min Luo
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
- Institute of Brain Sciences, School of Medicine, Jinan University, Guangzhou, 510632 China
- The Joint Laboratory of Brain Function and Health, Jinan University and The University of Hong Kong, Jinan University, Guangzhou, 510632 China
| |
Collapse
|
35
|
|
36
|
Neurotrophic effect of citrus 5-hydroxy-3,6,7,8,3',4'-hexamethoxyflavone: promotion of neurite outgrowth via cAMP/PKA/CREB pathway in PC12 cells. PLoS One 2011; 6:e28280. [PMID: 22140566 PMCID: PMC3226691 DOI: 10.1371/journal.pone.0028280] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 11/04/2011] [Indexed: 12/15/2022] Open
Abstract
5-Hydroxy-3,6,7,8,3′,4′-hexamethoxyflavone (5-OH-HxMF), a hydroxylated polymethoxyflavone, is found exclusively in the Citrus genus, particularly in the peels of sweet orange. In this research, we report the first investigation of the neurotrophic effects and mechanism of 5-OH-HxMF in PC12 pheochromocytoma cells. We found that 5-OH-HxMF can effectively induce PC12 neurite outgrowth accompanied with the expression of neuronal differentiation marker protein growth-associated protein-43(GAP-43). 5-OH-HxMF caused the enhancement of cyclic AMP response element binding protein (CREB) phosphorylation, c-fos gene expression and CRE-mediated transcription, which was inhibited by 2-naphthol AS-E phosphate (KG-501), a specific antagonist for the CREB-CBP complex formation. Moreover, 5-OH-HxMF-induced both CRE transcription activity and neurite outgrowth were inhibited by adenylate cyclase and protein kinase A (PKA) inhibitor, but not MEK1/2, protein kinase C (PKC), phosphatidylinositol 3-kinase (PI3K) or calcium/calmodulin-dependent protein kinase (CaMK) inhibitor. Consistently, 5-OH-HxMF treatment increased the intracellular cAMP level and downstream component, PKA activity. We also found that addition of K252a, a TrKA antagonist, significantly inhibited NGF- but not 5-OH-HxMF-induced neurite outgrowth. These results reveal for the first time that 5-OH-HxMF is an effective neurotrophic agent and its effect is mainly through a cAMP/PKA-dependent, but TrKA-independent, signaling pathway coupling with CRE-mediated gene transcription. A PKC-dependent and CREB-independent pathway was also involved in its neurotrophic action.
Collapse
|
37
|
Arregui L, Benítez JA, Razgado LF, Vergara P, Segovia J. Adenoviral astrocyte-specific expression of BDNF in the striata of mice transgenic for Huntington's disease delays the onset of the motor phenotype. Cell Mol Neurobiol 2011; 31:1229-43. [PMID: 21681558 DOI: 10.1007/s10571-011-9725-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Accepted: 06/03/2011] [Indexed: 10/18/2022]
Abstract
Huntington's disease (HD) is a neurodegenerative disorder characterized by motor, cognitive, and psychiatric symptoms. The most characteristic structural feature of this disease is neurodegeneration accompanied by gliosis in the striatum. BDNF has been proposed to protect striatal neurons from degeneration, because it is an important survival factor for these neurons from development to adulthood. Considering the extensive gliosis and the survival effects of BDNF, we constructed an adenovirus to express a BDNF cDNA in astrocyte cells using a promoter of the glial fibrillary acidic protein gene. Cells stably transfected in vitro with a BDNF cDNA driven by this promoter expressed BDNF and responded to external stimuli increasing BDNF production. When the vector was applied into the striata of mice transgenic for HD, long-term expression of the transgene was observed, associated with a delay of onset of the motor phenotype of the R6/2 HD transgenic mice. The present data indicate that the striatal expression of BDNF is a potential adjuvant for the treatment of HD.
Collapse
Affiliation(s)
- Leticia Arregui
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Av. Instituto Politécnico Nacional # 2508, 07360 Mexico, DF, Mexico
| | | | | | | | | |
Collapse
|
38
|
Jørgensen JR, Fransson A, Fjord-Larsen L, Thompson LH, Houchins JP, Andrade N, Torp M, Kalkkinen N, Andersson E, Lindvall O, Ulfendahl M, Brunak S, Johansen TE, Wahlberg LU. Cometin is a novel neurotrophic factor that promotes neurite outgrowth and neuroblast migration in vitro and supports survival of spiral ganglion neurons in vivo. Exp Neurol 2011; 233:172-81. [PMID: 21985865 DOI: 10.1016/j.expneurol.2011.09.027] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 08/31/2011] [Accepted: 09/20/2011] [Indexed: 12/22/2022]
Abstract
Neurotrophic factors are secreted proteins responsible for migration, growth and survival of neurons during development, and for maintenance and plasticity of adult neurons. Here we present a novel secreted protein named Cometin which together with Meteorin defines a new evolutionary conserved protein family. During early mouse development, Cometin is found exclusively in the floor plate and from E13.5 also in dorsal root ganglions and inner ear but apparently not in the adult nervous system. In vitro, Cometin promotes neurite outgrowth from dorsal root ganglion cells which can be blocked by inhibition of the Janus or MEK kinases. In this assay, additive effects of Cometin and Meteorin are observed indicating separate receptors. Furthermore, Cometin supports migration of neuroblasts from subventricular zone explants to the same extend as stromal cell derived factor 1a. Given the neurotrophic properties in vitro, combined with the restricted inner ear expression during development, we further investigated Cometin in relation to deafness. In neomycin deafened guinea pigs, two weeks intracochlear infusion of recombinant Cometin supports spiral ganglion neuron survival and function. In contrast to the control group receiving artificial perilymph, Cometin treated animals retain normal electrically-evoked brainstem response which is maintained several weeks after treatment cessation. Neuroprotection is also evident from stereological analysis of the spiral ganglion. Altogether, these studies show that Cometin is a potent new neurotrophic factor with therapeutic potential.
Collapse
|
39
|
Labhasetwar V. Neurodegenerative diseases: challenges. Drug Deliv Transl Res 2011; 1:349-50. [PMID: 25788420 DOI: 10.1007/s13346-011-0040-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- Vinod Labhasetwar
- Department of Biomedical Engineering/ND20, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA,
| |
Collapse
|
40
|
Fahnestock M. Brain-derived neurotrophic factor: the link between amyloid-β and memory loss. FUTURE NEUROLOGY 2011. [DOI: 10.2217/fnl.11.44] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is a critical molecule for learning and memory. Brain BDNF levels correlate with cognitive status. BDNF is downregulated in Alzheimer’s disease, in age-related cognitive impairment and in a variety of other neurodegenerative and psychiatric disorders exhibiting cognitive deficits. BDNF is downregulated in the Alzheimer’s disease brain by soluble, aggregated amyloid-β, acting via a pathway involving the transcription factor cAMP response element binding protein, which activates BDNF transcript IV. The complete pathway by which BDNF is downregulated is still unclear, and the diagnostic and therapeutic use of BDNF in neurodegenerative disease has not yet been exploited.
Collapse
Affiliation(s)
- Margaret Fahnestock
- Department of Psychiatry & Behavioural Neurosciences, McMaster University, 1280 Main Street West, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
41
|
Mazzio EA, Close F, Soliman KFA. The biochemical and cellular basis for nutraceutical strategies to attenuate neurodegeneration in Parkinson's disease. Int J Mol Sci 2011; 12:506-69. [PMID: 21340000 PMCID: PMC3039966 DOI: 10.3390/ijms12010506] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Revised: 01/05/2011] [Accepted: 01/14/2011] [Indexed: 12/19/2022] Open
Abstract
Future therapeutic intervention that could effectively decelerate the rate of degeneration within the substantia nigra pars compacta (SNc) could add years of mobility and reduce morbidity associated with Parkinson’s disease (PD). Neurodegenerative decline associated with PD is distinguished by extensive damage to SNc dopaminergic (DAergic) neurons and decay of the striatal tract. While genetic mutations or environmental toxins can precipitate pathology, progressive degenerative succession involves a gradual decline in DA neurotransmission/synaptic uptake, impaired oxidative glucose consumption, a rise in striatal lactate and chronic inflammation. Nutraceuticals play a fundamental role in energy metabolism and signaling transduction pathways that control neurotransmission and inflammation. However, the use of nutritional supplements to slow the progression of PD has met with considerable challenge and has thus far proven unsuccessful. This review re-examines precipitating factors and insults involved in PD and how nutraceuticals can affect each of these biological targets. Discussed are disease dynamics (Sections 1 and 2) and natural substances, vitamins and minerals that could impact disease processes (Section 3). Topics include nutritional influences on α-synuclein aggregation, ubiquitin proteasome function, mTOR signaling/lysosomal-autophagy, energy failure, faulty catecholamine trafficking, DA oxidation, synthesis of toxic DA-quinones, o-semiquinones, benzothiazolines, hyperhomocyseinemia, methylation, inflammation and irreversible oxidation of neuromelanin. In summary, it is clear that future research will be required to consider the multi-faceted nature of this disease and re-examine how and why the use of nutritional multi-vitamin-mineral and plant-based combinations could be used to slow the progression of PD, if possible.
Collapse
Affiliation(s)
- Elizabeth A Mazzio
- Florida A&M University, College of Pharmacy and Pharmaceutical Sciences, Tallahassee, FL 32307, USA; E-Mails: (E.A.M.); (F.C.)
| | | | | |
Collapse
|
42
|
Maher P, Dargusch R, Bodai L, Gerard PE, Purcell JM, Marsh JL. ERK activation by the polyphenols fisetin and resveratrol provides neuroprotection in multiple models of Huntington's disease. Hum Mol Genet 2010; 20:261-70. [PMID: 20952447 DOI: 10.1093/hmg/ddq460] [Citation(s) in RCA: 168] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Huntington's disease (HD) is an inherited, progressive and ultimately fatal neurodegenerative disorder that is characterized by psychiatric, cognitive and motor symptoms. Among the pathways implicated in HD are those involving mitogen-activated protein kinase signaling and particularly the Ras-extracellular signal-regulated kinase (ERK) cascade. Studies in both cells and animal models suggest that ERK activation might provide a novel therapeutic target for the treatment of HD but compounds that specifically activate ERK are few. To test the hypothesis that pharmaceutical activation of ERK might be protective for HD, a polyphenol, fisetin, which was previously shown to activate the Ras-ERK cascade, was tested in three different models of HD: PC12 cells expressing mutant Httex1 under the control of an inducible promoter, Drosophila expressing mutant Httex1 and the R6/2 mouse model of HD. The results indicate that fisetin can reduce the impact of mutant huntingtin in each of these disease models. Prompted by this observation, we determined that the related polyphenol, resveratrol, also activates ERK and is protective in HD models. Notably, although more than a dozen small molecule inhibitors of ERK activation are in clinical trials, very few small molecule activators of ERK signaling are reported. Thus, fisetin, resveratrol and related compounds might be useful for the treatment of HD by virtue of their unique ability to activate ERK.
Collapse
Affiliation(s)
- Pamela Maher
- Department of Cellular Neurobiology, The Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| | | | | | | | | | | |
Collapse
|
43
|
Nesti C, Pardini C, Barachini S, D'Alessandro D, Siciliano G, Murri L, Petrini M, Vaglini F. Human dental pulp stem cells protect mouse dopaminergic neurons against MPP+ or rotenone. Brain Res 2010; 1367:94-102. [PMID: 20854799 DOI: 10.1016/j.brainres.2010.09.042] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Revised: 09/08/2010] [Accepted: 09/11/2010] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the progressive death of substantia nigra dopaminergic neurons that results in a regional loss of striatal dopamine (DA) levels. Dental pulp contains ex vivo-expandable cells called dental pulp stem cells (DPSCs), with the capacity to differentiate into multiple cell lineages. More interestingly, due to their embryonic origin, DPSCs express neurotrophic factors such as brain-derived neurotrophic factor, nerve growth factor and glial cell-derived neurotrophic factor. The aim of the present study was to investigate the neuroprotective effects of DPSCs against MPP+ (2.5, 5, and 10 μM) and rotenone (0.25, 0.5 and 1 μM) in an in vitro model of PD, using an indirect co-culture system with mesencephalic cell cultures. When mesencephalic cultures were challenged with MPP+ or rotenone, in the presence of DPSCs a statistically significant protective effect was observed at all the tested doses in terms of DA uptake. DPSCs protective effect on DA neurons was also confirmed by immunocytochemistry: an increased number of spared tyrosine hydroxylase (TH)+ cells was observed in co-culture conditions compared to controls, and neurons showed longer processes in comparison with mesencephalic cells grown without DPSCs. In conclusion, the co-culture with DPSCs significantly attenuated MPP+ or rotenone-induced toxicity in primary cultures of mesencephalic neurons. Considering that the direct contact between the two cell types was prevented, it can be speculated that neuroprotection could be due to soluble factors such as BDNF and NGF, released by DPSCs. Blocking BDNF and NGF with neutralizing antibodies, the neuroprotecting effect of DPSCs was completely abolished. Therefore DPSCs can be viewed as possible candidates for studies on cell-based therapy in neurodegenerative disorders.
Collapse
Affiliation(s)
- Claudia Nesti
- RRMR/CUCCS (Rete Regionale di Medicina Rigenerativa/Center for the Clinical Use of Stem Cells), Italy; Stella Maris Scientific Institute, Pisa, Italy
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Lin CW, Wu MJ, Liu IYC, Su JD, Yen JH. Neurotrophic and cytoprotective action of luteolin in PC12 cells through ERK-dependent induction of Nrf2-driven HO-1 expression. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2010; 58:4477-4486. [PMID: 20302373 DOI: 10.1021/jf904061x] [Citation(s) in RCA: 117] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Luteolin (3',4',5,7-tetrahydroxyflavone), a food-derived flavonoid, has been reported to possess antioxidant, anti-inflammatory, and anticancer activities. In this work, we assessed whether luteolin has neurotrophic activity, namely, the ability to induce neurite outgrowth and to attenuate serum withdrawal-induced cytotoxicity in PC12 cells. Our results show that luteolin significantly induced neurite outgrowth along with increased expression of the differentiation marker, growth-associated protein-43 (GAP-43), in PC12 cells dose-dependently. Incubation of serum-deprived PC12 cells with luteolin prevented apoptosis, increased the expression of heme oxygenase-1 (HO-1) mRNA and protein levels, and enhanced the binding of nuclear factor E2-related factor 2 (Nrf2) to antioxidant response element (ARE), which works as an enhancer sequence in the HO-1 promoter. Addition of zinc protoporphyrin (Znpp), a selective HO-1 competitive inhibitor, significantly reduced the cytoprotective ability of luteolin, indicating the vital role of HO-1. Luteolin also persistently activated extracellular signal-regulated protein kinase 1/2 (ERK1/2); while the addition of U0126, a pharmacological MEK/ERK inhibitor, attenuated luteolin-induced Nrf2 binding activity, HO-1 expression, cytoprotective effect, and neurite outgrowth. Taken together, the above findings suggest that luteolin induces neurite outgrowth and augments cellular antioxidant defense capacity, at least in part, through the activation of the ERK signaling pathway.
Collapse
Affiliation(s)
- Chia-Wei Lin
- Department of Molecular Biology and Human Genetics, Tzu Chi University, Hualien, Taiwan
| | | | | | | | | |
Collapse
|
45
|
Li N, Liu GT. The novel squamosamide derivative FLZ enhances BDNF/TrkB/CREB signaling and inhibits neuronal apoptosis in APP/PS1 mice. Acta Pharmacol Sin 2010; 31:265-72. [PMID: 20154710 DOI: 10.1038/aps.2010.3] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM The aim of this study was to study the effects of compound FLZ, a novel cyclic derivative of squamosamide from Annona glabra, on brain-derived neurotrophic factor (BDNF)/tropomyosin receptor kinase B (TrkB)/cAMP response element-binding protein (CREB) signaling and neuronal apoptosis in the hippocampus of the amyloid precursor protein (APP)/presenilin-1 (PS1) double transgenic mice. METHODS APP/PS1 mice at the age of 5 months and age-matched wild-type mice (WT) were intragastrically administered FLZ (150 mg/kg) or vehicle [0.05% carboxymethyl cellulose sodium (CMC-Na)] daily for 20 weeks. The levels of BDNF in the hippocampus of WT and APP/PS1 mice were then measured by immunohistochemistry and Western blot analysis. Neuronal apoptosis in mouse hippocampus was detected by Nissl staining. Expression of NGF, NT3, pTrkB (Tyr515)/TrkB, pAkt (Ser473)/Akt, pERK/ERK, pCREB (Ser133)/CREB, Bcl-2/Bax, and active caspase-3 fragment/caspase-3 in the hippocampus of WT and APP/PS1 mice was detected by Western blot analysis. RESULTS Compared with vehicle-treated APP/PS1 mice, FLZ (150 mg/kg) significantly increased BDNF and NT3 expression in the hippocampus of APP/PS1 mice. In addition, FLZ promoted BDNF high-affinity receptor TrkB phosphorylation and activated its downstream ERK, thus increasing phosphorylation of CREB at Ser133 in the hippocampus of APP/PS1 mice. Moreover, FLZ showed neuroprotective effects on neuronal apoptosis by increasing the Bcl-2/Bax ratio and decreasing the active caspase-3 fragment/caspase-3 ratio in the hippocampus of APP/PS1 mice. CONCLUSION FLZ exerted neuroprotection at least partly through enhancing the BDNF/TrkB/CREB pathway and inhibiting neuronal apoptosis in APP/PS1 mice, which suggests that FLZ can be explored as a potential therapeutic agent in long-term Alzheimer's disease therapy.
Collapse
|
46
|
Bahat-Stroomza M, Barhum Y, Levy YS, Karpov O, Bulvik S, Melamed E, Offen D. Induction of adult human bone marrow mesenchymal stromal cells into functional astrocyte-like cells: potential for restorative treatment in Parkinson's disease. J Mol Neurosci 2009; 39:199-210. [PMID: 19127447 DOI: 10.1007/s12031-008-9166-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2008] [Accepted: 11/25/2008] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder with its motor phenomena due mostly to loss of dopamine-producing neurons in the substantia nigra. Pharmacological treatments aimed to increase the deficient dopaminergic neurotransmission are effective in ameliorating the cardinal symptoms, but none of these therapies is curative. It has been suggested that treatment with neurotrophic factors (NTFs) might protect and prevent death of the surviving dopaminergic neurons and induce proliferation of their axonal nerve terminals with reinnervations of the deafferented striatum. However, long-term delivery of such proteins into the CNS is problematic. We therefore aimed to differentiate ex vivo human bone marrow-derived mesenchymal stromal cells into astrocyte-like cells, capable of generating NTFs for future transplantation into basal ganglia of PD patients. Indeed, mesenchymal stromal cells treated with our novel astrocyte differentiation medium, present astrocyte-like morphology and express the astrocyte markers S100beta, glutamine synthetase and glial fibrillary acidic protein. Moreover, these astrocyte-like cells produce and secrete significant amounts of glial cell line-derived neurotrophic factor (GDNF), nerve growth factor (NGF), and brain-derived neurotrophic factor as indicated by messenger RNA, real-time polymerase chain reaction, ELISA, and Western blot analyses. Such NTF-producing cells transplanted into the striatum of 6-hydroxydopamine-lesioned rats, a model of PD, produced a progressive reduction in the apomorphine-induced contralateral rotations as well as behavioral improvement in rotor-rod and the "sunflower seeds" eating motor tests. Histological assessments revealed that the engrafted cells survived and expressed astrocyte and human markers and acted to regenerate the damaged dopaminergic nerve terminal system. Findings indicate that our novel procedure to induce NTF-producing astrocyte-like cells derived from human bone marrow stromal cells might become a promising and feasible autologous transplantation strategy for PD.
Collapse
Affiliation(s)
- Merav Bahat-Stroomza
- Laboratory of Neurosciences, Felsenstein Medical Research, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | | | | | | | |
Collapse
|
47
|
Mancuso C, Capone C, Ranieri SC, Fusco S, Calabrese V, Eboli ML, Preziosi P, Galeotti T, Pani G. Bilirubin as an endogenous modulator of neurotrophin redox signaling. J Neurosci Res 2008; 86:2235-49. [PMID: 18338802 DOI: 10.1002/jnr.21665] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Bilirubin is neurotoxic upon excess accumulation in the brain, but it also plays important physiological roles related to its antioxidant properties. Here we report that exposure of PC12 and primary rat cerebellar granule neurons to bilirubin (0.5-10 microM) drastically decreases nerve growth factor (NGF)/brain-derived neurotrophic factor signaling to Akt and extracellular signal-regulated kinases (ERKs), indicating a direct interference of the molecule with crucial prosurvival signaling pathways. This effect likely involves the scavenging capacity of bilirubin, the latter being able to inhibit, in PC12 cells, accumulation of intracellular reactive oxygen species and phosphorylation of Akt and ERKs in response to extracellular hydrogen peroxide. Interestingly, in the absence of exogenous growth factor, bilirubin elicited the phosphorylation of ERKs and of the cAMP responsive element binding (CREB) transcription factor, a signature of NGF-dependent survival signaling. These growth factor-like signaling effects were paralleled by the induction of the neuronal nitric oxide synthase (nNOS) and generation of nitric oxide (NO). Pharmacological dissection of the signaling cascade triggered by bilirubin revealed that phosphorylation of ERKs requires NO signaling through soluble guanylyl cyclase, and, further upstream, influx of extracellular calcium is necessary for nNOS induction and NO release, likely through calcium-dependent phosphorylation of CREB. Importantly, the cascade elicited by bilirubin through NO and ERK is cytoprotective, as revealed by exacerbated bilirubin toxicity in cultures treated by either NOS or MEK inhibitors. Taken together, these observations indicate an important action of bilirubin on redox signaling by neurotrophins, with either inhibitory or agonistic effects based on growth factor availability.
Collapse
Affiliation(s)
- Cesare Mancuso
- Institute of Pharmacology, Catholic University School of Medicine, Roma, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Saavedra A, Baltazar G, Duarte EP. Driving GDNF expression: the green and the red traffic lights. Prog Neurobiol 2008; 86:186-215. [PMID: 18824211 DOI: 10.1016/j.pneurobio.2008.09.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Revised: 06/18/2008] [Accepted: 09/03/2008] [Indexed: 01/28/2023]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is widely recognized as a potent survival factor for dopaminergic neurons of the nigrostriatal pathway that degenerate in Parkinson's disease (PD). In animal models of PD, GDNF delivery to the striatum or the substantia nigra protects dopaminergic neurons against subsequent toxin-induced injury and rescues previously damaged neurons, promoting recovery of the motor function. Thus, GDNF was proposed as a potential therapy to PD aimed at slowing down, halting or reversing neurodegeneration, an issue addressed in previous reviews. However, the use of GDNF as a therapeutic agent for PD is hampered by the difficulty in delivering it to the brain. Another potential strategy is to stimulate the endogenous expression of GDNF, but in order to do that we need to understand how GDNF expression is regulated. The aim of this review is to do a comprehensive analysis of the state of the art on the control of endogenous GDNF expression in the nervous system, focusing mainly on the nigrostriatal pathway. We address the control of GDNF expression during development, in the adult brain and after injury, and how damaged neurons signal glial cells to up-regulate GDNF. Pharmacological agents or natural molecules that increase GDNF expression and show neuroprotective activity in animal models of PD are reviewed. We also provide an integrated overview of the signalling pathways linking receptors for these molecules to the induction of GDNF gene, which might also become targets for neuroprotective therapies in PD.
Collapse
Affiliation(s)
- Ana Saavedra
- Department of Cell Biology, Immunology and Neurosciences, Faculty of Medicine, University of Barcelona, Carrer Casanova 143, 08036 Barcelona, Spain.
| | | | | |
Collapse
|
49
|
Signore AP, Zhang F, Weng Z, Gao Y, Chen J. Leptin neuroprotection in the CNS: mechanisms and therapeutic potentials. J Neurochem 2008; 106:1977-90. [PMID: 18466320 PMCID: PMC2634657 DOI: 10.1111/j.1471-4159.2008.05457.x] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Leptin is well known as a hormone important in the central control of appetitive behaviors via receptor-mediated actions in the hypothalamus, where leptin adjusts food intake to maintain homeostasis with the body's energy stores. Recent evidence has shown that leptin and its receptors are widespread in the CNS and may provide neuronal survival signals. This review summarizes our current knowledge of how leptin functions in the brain and then focuses on the ability of leptin to mitigate neuronal damage in experimental models of human neurological disorders. Damage to the brain by acute events such as stroke, or long-term loss of neurons associated with neurodegenerative diseases, including Parkinson's and Alzheimer's disease, may be amenable to treatment using leptin to limit death of susceptible cells. Leptin-mediated pro-survival signaling is now known to prevent the death of neurons in these models. The signaling cascades that leptin generates are shared by other neuroprotective molecules including insulin and erythropoietin, and are thus a component of the neurotrophic effects mediated by endogenous hormones. Coupled with evidence that leptin dysregulation in human disease also results in enhanced neuronal susceptibility to damage, development of leptin as a therapeutic methodology is an attractive and viable possibility.
Collapse
Affiliation(s)
- Armando P. Signore
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Feng Zhang
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Zhongfang Weng
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - YanQing Gao
- State Key Laboratory of Medical Neurobiology, Fudan University School of Medicine, Shanghai, China 200032
| | - Jun Chen
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
- State Key Laboratory of Medical Neurobiology, Fudan University School of Medicine, Shanghai, China 200032
- Geriatric Research, Educational and Clinical Center Veterans Affairs Pittsburgh Health Care System, Pittsburgh, Pennsylvania 15261
| |
Collapse
|
50
|
Apostol BL, Simmons DA, Zuccato C, Illes K, Pallos J, Casale M, Conforti P, Ramos C, Roarke M, Kathuria S, Cattaneo E, Marsh JL, Thompson LM. CEP-1347 reduces mutant huntingtin-associated neurotoxicity and restores BDNF levels in R6/2 mice. Mol Cell Neurosci 2008; 39:8-20. [PMID: 18602275 DOI: 10.1016/j.mcn.2008.04.007] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2008] [Revised: 03/27/2008] [Accepted: 04/11/2008] [Indexed: 01/09/2023] Open
Abstract
Huntington's disease (HD) is a devastating neurodegenerative disorder caused by an expanded polyglutamine repeat within the protein Huntingtin (Htt). We previously reported that mutant Htt expression activates the ERK1/2 and JNK pathways [Apostol, B.L., Illes, K., Pallos, J., Bodai, L., Wu, J., Strand, A., Schweitzer, E.S., Olson, J.M., Kazantsev, A., Marsh, J.L., Thompson, L.M., 2006. Mutant huntingtin alters MAPK signaling pathways in PC12 and striatal cells: ERK1/2 protects against mutant huntingtin-associated toxicity. Hum. Mol. Genet. 15, 273-285]. Chemical and genetic modulation of these pathways promotes cell survival and death, respectively. Here we test the ability of two closely related compounds, CEP-11004 and CEP-1347, which inhibit Mixed Lineage Kinases (MLKs) and are neuroprotective, to suppress mutant Htt-mediated pathogenesis in multiple model systems. CEP-11004/CEP-1347 treatment significantly decreased toxicity in mutant Htt-expressing cells that evoke a strong JNK response. However, suppression of cellular dysfunction in cell lines that exhibit only mild Htt-associated toxicity and little JNK activation was associated with activation of ERK1/2. These compounds also reduced neurotoxicity in immortalized striatal neurons from mutant knock-in mice and Drosophila expressing a mutant Htt fragment. Finally, CEP-1347 improved motor performance in R6/2 mice and restored expression of BDNF, a critical neurotrophic factor that is reduced in HD. These studies suggest a novel therapeutic approach for a currently untreatable neurodegenerative disease, HD, via CEP-1347 up-regulation of BDNF.
Collapse
Affiliation(s)
- Barbara L Apostol
- Department of Psychiatry and Human Behavior, University of California, Irvine, CA 92697, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|