1
|
Zhu Y, Hui Q, Zhang Z, Fu H, Qin Y, Zhao Q, Li Q, Zhang J, Guo L, He W, Han C. Advancements in the study of synaptic plasticity and mitochondrial autophagy relationship. J Neurosci Res 2024; 102:e25309. [PMID: 38400573 DOI: 10.1002/jnr.25309] [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: 10/09/2023] [Revised: 01/26/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024]
Abstract
Synapses serve as the points of communication between neurons, consisting primarily of three components: the presynaptic membrane, synaptic cleft, and postsynaptic membrane. They transmit signals through the release and reception of neurotransmitters. Synaptic plasticity, the ability of synapses to undergo structural and functional changes, is influenced by proteins such as growth-associated proteins, synaptic vesicle proteins, postsynaptic density proteins, and neurotrophic growth factors. Furthermore, maintaining synaptic plasticity consumes more than half of the brain's energy, with a significant portion of this energy originating from ATP generated through mitochondrial energy metabolism. Consequently, the quantity, distribution, transport, and function of mitochondria impact the stability of brain energy metabolism, thereby participating in the regulation of fundamental processes in synaptic plasticity, including neuronal differentiation, neurite outgrowth, synapse formation, and neurotransmitter release. This article provides a comprehensive overview of the proteins associated with presynaptic plasticity, postsynaptic plasticity, and common factors between the two, as well as the relationship between mitochondrial energy metabolism and synaptic plasticity.
Collapse
Affiliation(s)
- Yousong Zhu
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Qinlong Hui
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Zheng Zhang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Hao Fu
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Yali Qin
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Qiong Zhao
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Qinqing Li
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| | - Junlong Zhang
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
| | - Lei Guo
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
| | - Wenbin He
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
| | - Cheng Han
- Shanxi Key Laboratory of Chinese Medicine Encephalopathy, Jinzhong, China
- National International Joint Research Center for Molecular Traditional Chinese Medicine, Jinzhong, China
- Basic Medical College of Shanxi University of Chinese Medicine, Jinzhong, China
| |
Collapse
|
2
|
Kang Q, Mu Y, Yuan Y, Wang Y, Jin S, Wang C, Li Y. Diastereoselective Synthesis of Bicyclo[3.3.0]octenones by Copper-Catalyzed Transannular Ring-Closing Reaction. Org Lett 2022; 24:5924-5928. [PMID: 35930708 DOI: 10.1021/acs.orglett.2c02163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A novel and efficient copper-catalyzed transannular ring-closing reaction of eight-membered rings has been developed that provides a straightforward way to synthesize bicyclo[3.3.0]octane derivatives in good yields. Mechanistic studies revealed that the reaction pathway might involve chlorination followed by the Kornblum reaction. Readily accessible starting materials and good functional group tolerance make this procedure attractive.
Collapse
Affiliation(s)
- Qiongwen Kang
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Yuanyang Mu
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Yang Yuan
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Ye Wang
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Shuxin Jin
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| | - Chengyu Wang
- School of Chemistry and Chemical Engineering, Linyi University, Shuangling Road, Linyi, Shandong 276000, China
| | - Yanzhong Li
- School of Chemistry and Molecular Engineering, East China Normal University, 500 Dongchuan Road, Shanghai 200241, China
| |
Collapse
|
3
|
Boado RJ. IgG Fusion Proteins for Brain Delivery of Biologics via Blood-Brain Barrier Receptor-Mediated Transport. Pharmaceutics 2022; 14:pharmaceutics14071476. [PMID: 35890374 PMCID: PMC9322584 DOI: 10.3390/pharmaceutics14071476] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/10/2022] [Accepted: 07/12/2022] [Indexed: 01/01/2023] Open
Abstract
The treatment of neurological disorders with large-molecule biotherapeutics requires that the therapeutic drug be transported across the blood–brain barrier (BBB). However, recombinant biotherapeutics, such as neurotrophins, enzymes, decoy receptors, and monoclonal antibodies (MAb), do not cross the BBB. These biotherapeutics can be re-engineered as brain-penetrating bifunctional IgG fusion proteins. These recombinant proteins comprise two domains, the transport domain and the therapeutic domain, respectively. The transport domain is an MAb that acts as a molecular Trojan horse by targeting a BBB-specific endogenous receptor that induces receptor-mediated transcytosis into the brain, such as the human insulin receptor (HIR) or the transferrin receptor (TfR). The therapeutic domain of the IgG fusion protein exerts its pharmacological effect in the brain once across the BBB. A generation of bifunctional IgG fusion proteins has been engineered using genetically engineered MAbs directed to either the BBB HIR or TfR as the transport domain. These IgG fusion proteins were validated in animal models of lysosomal storage disorders; acute brain conditions, such as stroke; or chronic neurodegeneration, such as Parkinson’s disease and Alzheimer’s disease. Human phase I–III clinical trials were also completed for Hurler MPSI and Hunter MPSII using brain-penetrating IgG-iduronidase and -iduronate-2-sulfatase fusion protein, respectively.
Collapse
Affiliation(s)
- Ruben J Boado
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
4
|
Pardridge WM. A Historical Review of Brain Drug Delivery. Pharmaceutics 2022; 14:1283. [PMID: 35745855 PMCID: PMC9229021 DOI: 10.3390/pharmaceutics14061283] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
The history of brain drug delivery is reviewed beginning with the first demonstration, in 1914, that a drug for syphilis, salvarsan, did not enter the brain, due to the presence of a blood-brain barrier (BBB). Owing to restricted transport across the BBB, FDA-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Drugs that do not cross the BBB can be re-engineered for transport on endogenous BBB carrier-mediated transport and receptor-mediated transport systems, which were identified during the 1970s-1980s. By the 1990s, a multitude of brain drug delivery technologies emerged, including trans-cranial delivery, CSF delivery, BBB disruption, lipid carriers, prodrugs, stem cells, exosomes, nanoparticles, gene therapy, and biologics. The advantages and limitations of each of these brain drug delivery technologies are critically reviewed.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
5
|
Lee TK, Park JH, Ahn JH, Kim H, Song M, Lee JC, Kim JD, Jeon YH, Choi JH, Lee CH, Hwang IK, Yan BC, Won MH, Kang IJ. Pretreatment of Populus tomentiglandulosa protects hippocampal CA1 pyramidal neurons from ischemia-reperfusion injury in gerbils via increasing SODs expressions and maintaining BDNF and IGF-I expressions. Chin J Nat Med 2019; 17:424-434. [PMID: 31262455 DOI: 10.1016/s1875-5364(19)30050-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Indexed: 12/31/2022]
Abstract
To examine the effects of Populus tomentiglandulosa (PT) extract on the expressions of antioxidant enzymes and neurotrophic factors in the cornu ammonis 1 (CA1) region of the hippocampus at 5 min after inducing transient global cerebral ischemia (TGCI) in gerbils, TGCI was induced by occlusion of common carotid arteries for 5 min. Before ischemic surgery, 200 mg·kg-1 PT extract was orally administrated once daily for 7 d. We performed neuronal nuclear antigen immunohistochemistry and Fluoro-Jade B staining. Furthermore, we determined in situ production of superoxide anion radical, expression levels of SOD1 and SOD2 as antioxidant enzymes and brain-derived neurotrophic factor (BDNF) and insulin-like growth factor I (IGF-I) as neurotrophic factors. Pretreatment with 200 mg·kg-1 PT extract prevented neuronal death (loss). Furthermore, pretreatment with 200 mg·kg-1 PT extract significantly inhibited the production of superoxide anion radical, increased expressions of SODs and maintained expressions of BDNF and IGF-I. Such increased expressions of SODs were maintained in the neurons after IRI. In summary, pretreated PT extract can significantly increase levels of SODs and protect the neurons against TGCI, suggesting that PT can be a useful natural agent to protect against TGCI.
Collapse
Affiliation(s)
- Tae-Kyeong Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Joon Ha Park
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science, Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon 24252, Republic of Korea
| | - Hyunjung Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Minah Song
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Jong Dai Kim
- Division of Food Biotechnology, School of Biotechnology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Yong Hwan Jeon
- Department of Radiology, School of Medicine, Kangwon National University, Chuncheon 24289, Republic of Korea
| | - Jung Hoon Choi
- Department of Anatomy, College of Veterinary Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Choong Hyun Lee
- Department of Pharmacy, College of Pharmacy, Dankook University, Cheonan 31116, Republic of Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine and Research Institute for Veterinary Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Bing-Chun Yan
- Jiangsu Key Laboratory of Integrated Traditional Chinese, Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou 225001, China
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon 24341, Republic of Korea.
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon 24252, Republic of Korea.
| |
Collapse
|
6
|
Profiling withanolide A for therapeutic targets in neurodegenerative diseases. Bioorg Med Chem 2019; 27:2508-2520. [DOI: 10.1016/j.bmc.2019.03.022] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Revised: 03/08/2019] [Accepted: 03/10/2019] [Indexed: 11/22/2022]
|
7
|
Furtado D, Björnmalm M, Ayton S, Bush AI, Kempe K, Caruso F. Overcoming the Blood-Brain Barrier: The Role of Nanomaterials in Treating Neurological Diseases. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1801362. [PMID: 30066406 DOI: 10.1002/adma.201801362] [Citation(s) in RCA: 387] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/09/2018] [Indexed: 05/24/2023]
Abstract
Therapies directed toward the central nervous system remain difficult to translate into improved clinical outcomes. This is largely due to the blood-brain barrier (BBB), arguably the most tightly regulated interface in the human body, which routinely excludes most therapeutics. Advances in the engineering of nanomaterials and their application in biomedicine (i.e., nanomedicine) are enabling new strategies that have the potential to help improve our understanding and treatment of neurological diseases. Herein, the various mechanisms by which therapeutics can be delivered to the brain are examined and key challenges facing translation of this research from benchtop to bedside are highlighted. Following a contextual overview of the BBB anatomy and physiology in both healthy and diseased states, relevant therapeutic strategies for bypassing and crossing the BBB are discussed. The focus here is especially on nanomaterial-based drug delivery systems and the potential of these to overcome the biological challenges imposed by the BBB. Finally, disease-targeting strategies and clearance mechanisms are explored. The objective is to provide the diverse range of researchers active in the field (e.g., material scientists, chemists, engineers, neuroscientists, and clinicians) with an easily accessible guide to the key opportunities and challenges currently facing the nanomaterial-mediated treatment of neurological diseases.
Collapse
Affiliation(s)
- Denzil Furtado
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Mattias Björnmalm
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
- Department of Materials, Department of Bioengineering, and the Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ, UK
| | - Scott Ayton
- Melbourne Dementia Research Centre, The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, The Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, 3052, Australia
- Cooperative Research Center for Mental Health, Parkville, Victoria, 3052, Australia
| | - Kristian Kempe
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, 3052, Australia
| | - Frank Caruso
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, and the Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria, 3010, Australia
| |
Collapse
|
8
|
Simonato M. Neurotrophic factors and status epilepticus. Epilepsia 2018; 59 Suppl 2:87-91. [DOI: 10.1111/epi.14501] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/23/2017] [Indexed: 01/02/2023]
Affiliation(s)
- Michele Simonato
- Department of Medical Sciences; University of Ferrara; Ferrara Italy
- Division of Neuroscience; University Vita-Salute San Raffaele; Milan Italy
| |
Collapse
|
9
|
Delayed histochemical alterations within the neurovascular unit due to transient focal cerebral ischemia and experimental treatment with neurotrophic factors. PLoS One 2017; 12:e0174996. [PMID: 28445478 PMCID: PMC5405989 DOI: 10.1371/journal.pone.0174996] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2016] [Accepted: 03/17/2017] [Indexed: 02/03/2023] Open
Abstract
Current stroke therapy is focused on recanalizing strategies, but neuroprotective co-treatments are still lacking. Modern concepts of the ischemia-affected neurovascular unit (NVU) and surrounding penumbra emphasize the complexity during the transition from initial damaging to regenerative processes. While early treatment with neurotrophic factors was shown to result in lesion size reduction and blood-brain barrier (BBB) stabilization, cellular consequences from these treatments are poorly understood. This study explored delayed cellular responses not only to ischemic stroke, but also to an early treatment with neurotrophic factors. Rats underwent 60 minutes of focal cerebral ischemia. Fluorescence labeling was applied to sections from brains perfused 7 days after ischemia. Analyses focused on NVU constituents including the vasculature, astrocytes and microglia in the ischemic striatum, the border zone and the contralateral hemisphere. In addition to histochemical signs of BBB breakdown, a strong up-regulation of collagen IV and microglia activation occurred within the ischemic core with simultaneous degradation of astrocytes and their endfeet. Activated astroglia were mainly depicted at the border zone in terms of a glial scar formation. Early treatment with pigment epithelium-derived factor (PEDF) resulted in an attenuation of the usually up-regulated collagen IV-immunoreactivity. However, glial activation was not influenced by treatment with PEDF or the epidermal growth factor (EGF). In conclusion, these data on ischemia-induced cellular reactions within the NVU might help to develop treatments addressing the transition from injury towards regeneration. Thereby, the integrity of the vasculature in close relation to neighboring structures like astrocytes appears as a promising target.
Collapse
|
10
|
Kärkäs M, Porco JA, Stephenson CRJ. Photochemical Approaches to Complex Chemotypes: Applications in Natural Product Synthesis. Chem Rev 2016; 116:9683-747. [PMID: 27120289 PMCID: PMC5025835 DOI: 10.1021/acs.chemrev.5b00760] [Citation(s) in RCA: 701] [Impact Index Per Article: 77.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Indexed: 01/29/2023]
Abstract
The use of photochemical transformations is a powerful strategy that allows for the formation of a high degree of molecular complexity from relatively simple building blocks in a single step. A central feature of all light-promoted transformations is the involvement of electronically excited states, generated upon absorption of photons. This produces transient reactive intermediates and significantly alters the reactivity of a chemical compound. The input of energy provided by light thus offers a means to produce strained and unique target compounds that cannot be assembled using thermal protocols. This review aims at highlighting photochemical transformations as a tool for rapidly accessing structurally and stereochemically diverse scaffolds. Synthetic designs based on photochemical transformations have the potential to afford complex polycyclic carbon skeletons with impressive efficiency, which are of high value in total synthesis.
Collapse
Affiliation(s)
- Markus
D. Kärkäs
- Department
of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, Michigan 48109, United States
| | - John A. Porco
- Department
of Chemistry, Center for Molecular Discovery (BU-CMD), Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States
| | - Corey R. J. Stephenson
- Department
of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
11
|
Voutilainen MH, Arumäe U, Airavaara M, Saarma M. Therapeutic potential of the endoplasmic reticulum located and secreted CDNF/MANF family of neurotrophic factors in Parkinson's disease. FEBS Lett 2015; 589:3739-48. [PMID: 26450777 DOI: 10.1016/j.febslet.2015.09.031] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/23/2015] [Accepted: 09/30/2015] [Indexed: 12/16/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder where dopamine (DA) neurons in the substantia nigra degenerate and die. Since no cure for PD exists, there is a need for disease-modifying drugs. Glial cell line-derived neurotrophic factor (GDNF) and related neurturin (NRTN) can protect and repair DA neurons in neurotoxin animal models of PD. However, GDNF was unable to rescue DA neurons in an α-synuclein model of PD, and both factors have shown modest effects in phase two clinical trials. Neurotrophic factors (NTFs), cerebral DA NTF (CDNF) and mesencephalic astrocyte-derived NTF (MANF) form a novel family of evolutionarily conserved, endoplasmic reticulum (ER) located and secreted NTFs. CDNF and MANF have a unique structure and an unparalleled dual mode of action that differs from other known NTFs. Both protect cells from ER stress, and regulate the unfolded protein response via interacting with chaperons, and CDNF dissolves intracellular α-synuclein aggregates. By binding to putative plasma membrane receptors, they promote the survival of DA neurons similarly to conventional NTFs. In animal models of PD, CDNF protects and repairs DA neurons, regulates ER stress, and improves motor function more efficiently than other NTFs.
Collapse
Affiliation(s)
| | - Urmas Arumäe
- Institute of Biotechnology, University of Helsinki, Finland; Department of Gene Technology, Tallinn University of Technology, Tallinn, Estonia
| | | | - Mart Saarma
- Institute of Biotechnology, University of Helsinki, Finland.
| |
Collapse
|
12
|
|
13
|
Menstrual Blood Transplantation Therapy for Stroke and Other Neurological Disorders. Regen Med 2015. [DOI: 10.1007/978-1-4471-6542-2_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
14
|
Nakada M. Enantioselective Total Syntheses of Cyathane Diterpenoids. CHEM REC 2014; 14:641-62. [DOI: 10.1002/tcr.201402019] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Indexed: 11/09/2022]
Affiliation(s)
- Masahisa Nakada
- Department of Chemistry and Biochemistry; School of Advanced Science and Engineering; Waseda University; 3-4-1 Ohkubo, Shinjuku-ku Tokyo 169-8555 Japan
| |
Collapse
|
15
|
Upadhyay RK. Drug delivery systems, CNS protection, and the blood brain barrier. BIOMED RESEARCH INTERNATIONAL 2014; 2014:869269. [PMID: 25136634 PMCID: PMC4127280 DOI: 10.1155/2014/869269] [Citation(s) in RCA: 253] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 05/31/2014] [Accepted: 06/05/2014] [Indexed: 12/12/2022]
Abstract
Present review highlights various drug delivery systems used for delivery of pharmaceutical agents mainly antibiotics, antineoplastic agents, neuropeptides, and other therapeutic substances through the endothelial capillaries (BBB) for CNS therapeutics. In addition, the use of ultrasound in delivery of therapeutic agents/biomolecules such as proline rich peptides, prodrugs, radiopharmaceuticals, proteins, immunoglobulins, and chimeric peptides to the target sites in deep tissue locations inside tumor sites of brain has been explained. In addition, therapeutic applications of various types of nanoparticles such as chitosan based nanomers, dendrimers, carbon nanotubes, niosomes, beta cyclodextrin carriers, cholesterol mediated cationic solid lipid nanoparticles, colloidal drug carriers, liposomes, and micelles have been discussed with their recent advancements. Emphasis has been given on the need of physiological and therapeutic optimization of existing drug delivery methods and their carriers to deliver therapeutic amount of drug into the brain for treatment of various neurological diseases and disorders. Further, strong recommendations are being made to develop nanosized drug carriers/vehicles and noninvasive therapeutic alternatives of conventional methods for better therapeutics of CNS related diseases. Hence, there is an urgent need to design nontoxic biocompatible drugs and develop noninvasive delivery methods to check posttreatment clinical fatalities in neuropatients which occur due to existing highly toxic invasive drugs and treatment methods.
Collapse
Affiliation(s)
- Ravi Kant Upadhyay
- Department of Zoology, DDU Gorakhpur University, Gorakhpur 273009, India
| |
Collapse
|
16
|
Viola TW, Tractenberg SG, Levandowski ML, Pezzi JC, Bauer ME, Teixeira AL, Grassi-Oliveira R. Neurotrophic factors in women with crack cocaine dependence during early abstinence: the role of early life stress. J Psychiatry Neurosci 2014; 39:206-14. [PMID: 24331739 PMCID: PMC3997606 DOI: 10.1503/jpn.130027] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 06/26/2013] [Accepted: 10/11/2013] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Neurotrophic factors have been investigated in the pathophysiology of alcohol and drug dependence and have been related to early life stress driving developmental programming of neuroendocrine systems. METHODS We conducted a follow-up study that aimed to assess the plasma levels of glial cell line-derived neurotrophic factor (GDNF), brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF), neurotrophin-3 (NT3) and neurotrophin-4/5 (NT4/5) in crack users during 3 weeks of early abstinence in comparison with healthy controls. We performed a comprehensive clinical assessment in female inpatients with crack cocaine dependence (separated into 2 groups: participants with (CSA+) and without (CSA-) a history of childhood sexual abuse) and a group of nonuser control participants. RESULTS Our sample included 104 women with crack cocaine dependence and 22 controls; of the women who used crack cocaine, 22 had a history of childhood sexual abuse and 82 did not. The GDNF plasma levels in the CSA+ group increased dramatically during 3 weeks of detoxification. In contrast, those in the CSA- group showed lower and stable levels of GDNF under the same conditions. Compared with the control group, BDNF plasma levels remained elevated and NGF levels were reduced during early abstinence. We found no differences in NT3 and NT4/5 between the patients and controls. However, within-group analyses showed that the CSA+ group exhibited higher levels of NT4/5 than the CSA- group at the end of detoxification. LIMITATIONS Some of the participants were using neuroleptics, mood stabilizers or antidepressants; our sample included only women; memory bias could not be controlled; and we did not investigate the possible confounding effects of other forms of stress during childhood. CONCLUSION This study supports the association between early life stress and peripheral neurotrophic factor levels in crack cocaine users. During early abstinence, plasmastic GDNF and NT4/5 were the only factors to show changes associated with a history of childhood sexual abuse.
Collapse
Affiliation(s)
- Thiago Wendt Viola
- Viola, Tractenberg, Levandowski, Bauer, Grassi-Oliveira — Centre of Studies and Research in Traumatic Stress, PostGraduate Program in Psychology, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Pezzi — Post-Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Bauer, Grassi-Oliveira — Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Teixeira — Neuroscience Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Saulo Gantes Tractenberg
- Viola, Tractenberg, Levandowski, Bauer, Grassi-Oliveira — Centre of Studies and Research in Traumatic Stress, PostGraduate Program in Psychology, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Pezzi — Post-Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Bauer, Grassi-Oliveira — Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Teixeira — Neuroscience Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Mateus Luz Levandowski
- Viola, Tractenberg, Levandowski, Bauer, Grassi-Oliveira — Centre of Studies and Research in Traumatic Stress, PostGraduate Program in Psychology, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Pezzi — Post-Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Bauer, Grassi-Oliveira — Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Teixeira — Neuroscience Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Júlio Carlos Pezzi
- Viola, Tractenberg, Levandowski, Bauer, Grassi-Oliveira — Centre of Studies and Research in Traumatic Stress, PostGraduate Program in Psychology, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Pezzi — Post-Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Bauer, Grassi-Oliveira — Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Teixeira — Neuroscience Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Moisés Evandro Bauer
- Viola, Tractenberg, Levandowski, Bauer, Grassi-Oliveira — Centre of Studies and Research in Traumatic Stress, PostGraduate Program in Psychology, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Pezzi — Post-Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Bauer, Grassi-Oliveira — Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Teixeira — Neuroscience Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Antonio Lúcio Teixeira
- Viola, Tractenberg, Levandowski, Bauer, Grassi-Oliveira — Centre of Studies and Research in Traumatic Stress, PostGraduate Program in Psychology, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Pezzi — Post-Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Bauer, Grassi-Oliveira — Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Teixeira — Neuroscience Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Rodrigo Grassi-Oliveira
- Viola, Tractenberg, Levandowski, Bauer, Grassi-Oliveira — Centre of Studies and Research in Traumatic Stress, PostGraduate Program in Psychology, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Pezzi — Post-Graduate Program in Health Sciences, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil; Bauer, Grassi-Oliveira — Laboratory of Immunosenescence, Institute of Biomedical Research, Pontifical Catholic University of the Rio Grande do Sul (PUCRS), Porto Alegre, Brazil; Teixeira — Neuroscience Program, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
17
|
Yang Y, Haskins CW, Zhang W, Low PL, Dai M. Divergent total syntheses of lyconadins A and C. Angew Chem Int Ed Engl 2014; 53:3922-5. [PMID: 24596132 PMCID: PMC4113559 DOI: 10.1002/anie.201400416] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Indexed: 12/13/2022]
Abstract
Divergent and concise total syntheses of two lycopodium alkaloids, lyconadins A and C have been developed. The synthesis of lyconadin A, having potent neurotrophic activity, features an efficient one-pot ketal removal and formal aza-[4+2] cyclization to form the cagelike core structure. A tandem ketal removal/Mannich reaction was developed to build the tricyclic structure of lyconadin C. Both lyconadins A and C were synthesized from a pivotal intermediate.
Collapse
Affiliation(s)
- Yang Yang
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907 (USA), Homepage: http://www.chem.purdue.edu/dai/
| | - Christopher W. Haskins
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907 (USA), Homepage: http://www.chem.purdue.edu/dai/
| | - Wandi Zhang
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907 (USA), Homepage: http://www.chem.purdue.edu/dai/
| | - Pui Leng Low
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907 (USA), Homepage: http://www.chem.purdue.edu/dai/
| | - Mingji Dai
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47907 (USA), Homepage: http://www.chem.purdue.edu/dai/
| |
Collapse
|
18
|
Yang Y, Haskins CW, Zhang W, Low PL, Dai M. Divergent Total Syntheses of Lyconadins A and C. Angew Chem Int Ed Engl 2014. [DOI: 10.1002/ange.201400416] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
19
|
Paradiso B, Zucchini S, Simonato M. Implication of fibroblast growth factors in epileptogenesis-associated circuit rearrangements. Front Cell Neurosci 2013; 7:152. [PMID: 24062643 PMCID: PMC3772316 DOI: 10.3389/fncel.2013.00152] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Accepted: 08/26/2013] [Indexed: 12/26/2022] Open
Abstract
The transformation of a normal brain in epileptic (epileptogenesis) is associated with extensive morpho-functional alterations, including cell death, axonal and dendritic plasticity, neurogenesis, and others. Neurotrophic factors (NTFs) appear to be very strongly implicated in these phenomena. In this review, we focus on the involvement of fibroblast growth factor (FGF) family members. Available data demonstrate that the FGFs are highly involved in the generation of the morpho-functional alterations in brain circuitries associated with epileptogenesis. For example, data on FGF2, the most studied member, suggest that it may be implicated both in seizure susceptibility and in seizure-induced plasticity, exerting different, and apparently contrasting effects: favoring acute seizures but reducing seizure-induced cell death. Even if many FGF members are still unexplored and very limited information is available on the FGF receptors, a complex and fascinating picture is emerging: multiple FGFs producing synergic or antagonistic effects one with another (and/or with other NTFs) on biological parameters that, in turn, facilitate or oppose transformation of the normal tissue in epileptic. In principle, identifying key elements in these phenomena may lead to effective therapies, but reaching this goal will require confronting a huge complexity. One first step could be to generate a "neurotrophicome" listing the FGFs (and all other NTFs) that are active during epileptogenesis. This should include identification of the extent to which each NTF is active (concentrations at the site of action); how it is active (local representation of receptor subtypes); when in the natural history of disease this occurs; how the NTF at hand will possibly interact with other NTFs. This is extraordinarily challenging, but holds the promise of a better understanding of epileptogenesis and, at large, of brain function.
Collapse
Affiliation(s)
- Beatrice Paradiso
- 1Department of Medical Sciences, Section of Pharmacology, University of Ferrara Ferrara, Italy ; 2Department of Morphology, Surgery and Experimental Medicine, Section of Pathology Ferrara, Italy ; 3National Institute of Neuroscience, University of Ferrara Ferrara, Italy
| | | | | |
Collapse
|
20
|
Kobayakawa Y, Nakada M. Total Syntheses of (−)-Scabronines G and A, and (−)-Episcabronine A. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201303224] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
21
|
Kobayakawa Y, Nakada M. Total Syntheses of (−)-Scabronines G and A, and (−)-Episcabronine A. Angew Chem Int Ed Engl 2013; 52:7569-73. [DOI: 10.1002/anie.201303224] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Indexed: 11/08/2022]
|
22
|
Abstract
INTRODUCTION Parkinson's disease (PD) is a common and chronic movement disorder with no therapy yet proven to alter the underlying advancing pathology. Gene delivery of trophic factors, which have shown disease modifying potential in preclinical PD models, are now being evaluated in early clinical trials. AREAS COVERED This review discusses early experiences with glial-derived neurotrophic factor in PD, the initial studies using AAV2-neurturin in PD patients, the lessons learned from these studies and the future directions of this therapy. EXPERT OPINION Gene therapy has emerged as a potential breakthrough in the treatment of PD and early clinical trials using AAV2-neurturin, a trophic factor that has shown the ability to protect dopaminergic degeneration in preclinical PD models, are underway. While trophic protection of dopamine neurons would be a significant breakthrough, PD remains a widespread disorder that involves neurodegeneration across multiple cellular types. We believe that these initial studies with AAV2-neurturin are significant steps toward the realization of gene delivery of trophic factors as a viable therapy, though the ultimate goal must be that of comprehensive neurorestoration.
Collapse
Affiliation(s)
- Patrick Hickey
- Duke University Medical Center, DUMC Box 3333, Durham, NC 27710, USA
| | | |
Collapse
|
23
|
Trzoss L, Xu J, Lacoske MH, Mobley WC, Theodorakis EA. Illicium sesquiterpenes: divergent synthetic strategy and neurotrophic activity studies. Chemistry 2013; 19:6398-408. [PMID: 23526661 PMCID: PMC3875175 DOI: 10.1002/chem.201300198] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Indexed: 01/12/2023]
Abstract
Majucin-type sesquiterpenes from Illicium sp., such as jiadifenolide (2), jiadifenin (3), and (1R,10S)-2-oxo-3,4-dehydroxyneomajucin (4, ODNM), possess a complex caged chemical architecture and remarkable neurotrophic activities. As such, they represent attractive small-molecule leads against various neurodegenerative diseases. We present an efficient, enantioselective, and unified synthesis of 2, 3, and 4 and designed analogues that diverge from tetracyclic key intermediate 7. The synthesis of 7 is highlighted by the use of an enantioselective Robinson annulation reaction (construction of the AB rings), a Pd-mediated carbomethoxylation reaction (construction of the C ring), and a one-pot oxidative reaction cascade (construction of the D ring). Evaluation of the neurotrophic activity of these compounds led to the identification of several highly potent small molecules that significantly enhanced the activity of nerve growth factor (NGF) in PC-12 cells. Moreover, efforts to define the common pharmacophoric motif suggest that substitution at the C-10 center significantly affects bioactivity, while the hemiketal moiety of 2 and 3 and the C-1 substitution might not be critical to the neurotrophic activity.
Collapse
Affiliation(s)
- Lynnie Trzoss
- Department of Chemistry and Biochemistry, University of California at San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0358, USA, Fax: (+)1-858-822-0386
| | - Jing Xu
- Department of Chemistry and Biochemistry, University of California at San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0358, USA, Fax: (+)1-858-822-0386
| | - Michelle H. Lacoske
- Department of Chemistry and Biochemistry, University of California at San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0358, USA, Fax: (+)1-858-822-0386
| | - William C. Mobley
- Department of Neurosciences, University of California at San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0752, USA
| | - Emmanuel A. Theodorakis
- Department of Chemistry and Biochemistry, University of California at San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0358, USA, Fax: (+)1-858-822-0386
| |
Collapse
|
24
|
Neurovascular protection by targeting early blood-brain barrier disruption with neurotrophic factors after ischemia-reperfusion in rats*. J Cereb Blood Flow Metab 2013; 33:557-66. [PMID: 23299242 PMCID: PMC3618392 DOI: 10.1038/jcbfm.2012.201] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The 'new penumbra' concept imbues the transition between injury and repair at the neurovascular unit with profound implications for selecting the appropriate type and timing of neuroprotective interventions. In this conceptual study, we investigated the protective effects of pigment epithelium-derived factor (PEDF) and compared them with the properties of epidermal growth factor (EGF) in a rat model of ischemia-reperfusion injury. We initiated a delayed intervention 3 hours after reperfusion using equimolar amounts of PEDF and EGF. These agents were then administered intravenously for 4 hours following reperfusion after 1 hour of focal ischemia. Magnetic resonance imaging indices were characterized, and imaging was performed at multiple time points post reperfusion. PEDF and EGF reduced lesion volumes at all time points as observed on T2-weighted images (T2-LVs). In addition PEDF selectively attenuated lesion volume expansion at 48 hours after reperfusion and persistently modulated blood-brain barrier (BBB) permeability at all time points. Intervention with peptides is suspected to cause edema formation at distant regions. The observed T2-LV reduction and BBB modulation by these trophic factors is probably mediated through a number of diverse mechanisms. A thorough evaluation of neurotrophins is still necessary to determine their time-dependent contributions against injury and their modulatory effects on repair after stroke.
Collapse
|
25
|
RODRIGUES MARIACAROLINAO, DMITRIEV DMITRIY, RODRIGUES ANTONIO, GLOVER LORENE, SANBERG PAULR, ALLICKSON JULIEG, KUZMIN-NICHOLS NICOLE, TAJIRI NAOKI, SHINOZUKA KAZUTAKA, GARBUZOVA-DAVIS SVITLANA, KANEKO YUJI, BORLONGAN CESARV. Menstrual blood transplantation for ischemic stroke: Therapeutic mechanisms and practical issues. Interv Med Appl Sci 2012; 4:59-68. [PMID: 25267932 PMCID: PMC4177033 DOI: 10.1556/imas.4.2012.2.1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Cerebrovascular diseases are a major cause of death and long-term disability in developed countries. Tissue plasmin activator (tPA) is the only approved therapy for ischemic stroke, strongly limited by the short therapeutic window and hemorrhagic complications, therefore excluding most patients from its benefits. The rescue of the penumbra area of the ischemic infarct is decisive for functional recovery after stroke. Inflammation is a key feature in the penumbra area and it plays a dual role, improving injury in early phases but impairing neural survival at later stages. Stem cells can be opportunely used to modulate inflammation, abrogate cell death and, therefore, preserve neural function. We here discuss the possible role of stem cells derived from menstrual blood as restorative treatment for stroke. We highlight the availability, proliferative capacity, pluripotentiality and angiogenic features of these cells and explore their present and future experimental and clinical applications.
Collapse
Affiliation(s)
- MARIA CAROLINA O. RODRIGUES
- Department of Neurosurgery and Brain Repair, University of South Florida, College of Medicine, Tampa, FL, USA
- Department of Internal Medicine, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - DMITRIY DMITRIEV
- Department of Neurosurgery and Brain Repair, University of South Florida, College of Medicine, Tampa, FL, USA
| | - ANTONIO RODRIGUES
- Department of Neurosurgery and Brain Repair, University of South Florida, College of Medicine, Tampa, FL, USA
- Department of Internal Medicine, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - LOREN E. GLOVER
- Department of Neurosurgery and Brain Repair, University of South Florida, College of Medicine, Tampa, FL, USA
| | - PAUL R. SANBERG
- Department of Neurosurgery and Brain Repair, University of South Florida, College of Medicine, Tampa, FL, USA
| | | | | | - NAOKI TAJIRI
- Department of Neurosurgery and Brain Repair, University of South Florida, College of Medicine, Tampa, FL, USA
| | - KAZUTAKA SHINOZUKA
- Department of Neurosurgery and Brain Repair, University of South Florida, College of Medicine, Tampa, FL, USA
| | - SVITLANA GARBUZOVA-DAVIS
- Department of Neurosurgery and Brain Repair, University of South Florida, College of Medicine, Tampa, FL, USA
| | - YUJI KANEKO
- Department of Neurosurgery and Brain Repair, University of South Florida, College of Medicine, Tampa, FL, USA
| | - CESAR V. BORLONGAN
- Department of Neurosurgery and Brain Repair, University of South Florida, College of Medicine, Tampa, FL, USA
| |
Collapse
|
26
|
Tanaka Y, Fukumitsu H, Soumiya H, Yoshimura S, Iwama T, Furukawa S. 2-decenoic acid ethyl ester, a compound that elicits neurotrophin-like intracellular signals, facilitating functional recovery from cerebral infarction in mice. Int J Mol Sci 2012; 13:4968-4981. [PMID: 22606023 PMCID: PMC3344259 DOI: 10.3390/ijms13044968] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 04/06/2012] [Accepted: 04/11/2012] [Indexed: 02/07/2023] Open
Abstract
In our previous study, we found that trans-2-decenoic acid ethyl ester (DAEE), a derivative of a medium-chain fatty acid, elicits neurotrophin-like signals including the activation of extracellular signal-regulated kinases 1 and 2 (ERK1/2) in cultured mouse cortical neurons. Here, we examined the efficacy of intraperitoneal administration of DAEE on the treatment of a mouse model of the cerebral infarction caused by unilateral permanent middle cerebral artery occlusion (PMCAO). DAEE-treatment (100 μg/kg body weight injected at 0.5, 24, 48, 72 h after PMCAO) significantly restored the mice from PMCAO-induced neurological deficits including motor paralysis when evaluated 48, 72, and 96 h after the PMCAO. Furthermore, DAEE facilitated the phosphorylation of ERK1/2 on the infarction side of the brain when analyzed by Western immunoblot analysis, and it enhanced the number of phosphorylated ERK1/2-positive cells in the border areas between the infarction and non-infarction regions of the cerebral cortex, as estimated immunohistochemically. As the infarct volume remained unchanged after DAEE-treatment, it is more likely that DAEE improved the neurological condition through enhanced neuronal functions of the remaining neurons in the damaged areas rather than by maintaining neuronal survival. These results suggest that DAEE has a neuro-protective effect on cerebral infarction.
Collapse
Affiliation(s)
- Yoshitaka Tanaka
- Laboratory of Molecular Biology, Gifu Pharmaceutical University, Daigaku-nishi, 1-25-4, Gifu 501-1190, Japan; E-Mails: (Y.T.); (H.F.): (H.S.)
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Yanagido 1-1, Gifu 501-1194, Japan; E-Mails: (S.Y.); (T.I.)
| | - Hidefumi Fukumitsu
- Laboratory of Molecular Biology, Gifu Pharmaceutical University, Daigaku-nishi, 1-25-4, Gifu 501-1190, Japan; E-Mails: (Y.T.); (H.F.): (H.S.)
| | - Hitomi Soumiya
- Laboratory of Molecular Biology, Gifu Pharmaceutical University, Daigaku-nishi, 1-25-4, Gifu 501-1190, Japan; E-Mails: (Y.T.); (H.F.): (H.S.)
| | - Shinichi Yoshimura
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Yanagido 1-1, Gifu 501-1194, Japan; E-Mails: (S.Y.); (T.I.)
| | - Toru Iwama
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Yanagido 1-1, Gifu 501-1194, Japan; E-Mails: (S.Y.); (T.I.)
| | - Shoei Furukawa
- Laboratory of Molecular Biology, Gifu Pharmaceutical University, Daigaku-nishi, 1-25-4, Gifu 501-1190, Japan; E-Mails: (Y.T.); (H.F.): (H.S.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +81-58-230-8100; Fax: +81-58-230-8105
| |
Collapse
|
27
|
Katsari E, Zikos C, Tziveleka LA, Paravatou-Petsotas M, Paleos CM. Cholesteryl-functionalized ADNF-9 peptide: enhanced membrane transport through mouse neuroblastoma Neuro-2a cells. Chem Biol Drug Des 2012; 80:148-54. [PMID: 22416980 DOI: 10.1111/j.1747-0285.2012.01381.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A cholesteryl-functionalized derivative of activity dependent neurotrophic factor-9 peptide (a nine amino acid core peptide of activity-dependent neurotrophic factor, acting against Alzheimer's disease) was synthesized aiming at the improvement of its bioavailability. Therefore, its uptake was comparatively investigated with that of its parent peptide by employing mouse neuroblastoma Neuro-2a cells. Owing to the hydrophobic character of this cholesteryl-functionalized peptide, it exhibited enhanced permeability and intracellular uptake while it also retained its low cytotoxicity at concentrations up to 1 μM. FACS analysis also revealed that when Neuro-2a cells were treated with this activity dependent neurotrophic factor-9 derivative, at a concentration of 50 nM, an almost 100% uptake was obtained. In addition, in vitro biological activity experiments showed that the functionalized peptide retained its neurotrophic activity at femtomolar concentration range.
Collapse
|
28
|
Trzoss L, Xu J, Lacoske MH, Mobley WC, Theodorakis EA. Enantioselective synthesis of (-)-jiadifenin, a potent neurotrophic modulator. Org Lett 2011; 13:4554-7. [PMID: 21812392 DOI: 10.1021/ol201742j] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The first enantioselective synthesis of (-)-jiadifenin (1), a potent neurite outgrowth promoter isolated from the Illicium species, is described. The synthetic strategy builds upon bicyclic motif 6, which represents the AB ring of the natural product and proceeds in 19 steps and 1.1% overall yield. Key to our approach is a Mn(III)-mediated oxidation reaction of A ring that, following a regio- and diastereoselective α-hydroxylation and methylation sequence, produces the desired functionalities of (-)-jiadifenin. The effect of synthetic 1 in NGF-mediated neurite outgrowth was also measured in PC-12 cells.
Collapse
Affiliation(s)
- Lynnie Trzoss
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California 92093-0358, USA
| | | | | | | | | |
Collapse
|
29
|
Bartus RT, Brown L, Wilson A, Kruegel B, Siffert J, Johnson EM, Kordower JH, Herzog CD. Properly scaled and targeted AAV2-NRTN (neurturin) to the substantia nigra is safe, effective and causes no weight loss: support for nigral targeting in Parkinson's disease. Neurobiol Dis 2011; 44:38-52. [PMID: 21704161 DOI: 10.1016/j.nbd.2011.05.026] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2011] [Revised: 05/06/2011] [Accepted: 05/28/2011] [Indexed: 10/18/2022] Open
Abstract
Recent analyses of autopsied brains from subjects previously administered AAV2-neurturin (NRTN) gene transfer argues that optimizing the effects of neurotrophic factors in Parkinson's disease (PD) likely requires delivery to both the degenerating cell bodies (in substantia nigra) and their terminals (in striatum). Prior to implementing this novel dosing paradigm in humans, we conducted eight nonclinical experiments with three general objectives: (1) evaluate the feasibility, safety and effectiveness of targeting the substantia nigra (SN) with AAV2-NRTN, (2) better understand and appraise recent warnings of serious weight loss that might occur with targeting the SN with neurotrophic factors, and (3) define an appropriate dose of AAV2-NRTN that should safely and effectively cover the SN in PD patients. Toward these ends, we first determined SN volume for rats, monkeys and humans, and employed these values to calculate comparable dose equivalents for each species by scaling each dose, based on relative SN volume. Using this information, we next injected AAV2-GFP to monkey SN to quantify AAV2-vector distribution and confirm reasonable SN coverage. We then selected and administered a ~200-fold range of AAV2-NRTN doses (and a single AAV2-GDNF dose) to rat SN, producing a wide range of protein expression. In contrast to recent warnings regarding nigra targeting, no dose produced any serious side effects or toxicity, though we replicated the modest reduction in weight gain reported by others with the highest AAV2-NRTN and the AAV2-GDNF dose. A dose-related increase in NRTN expression was seen, with the lower doses limiting NRTN to the peri-SN and the highest dose producing mistargeted NRTN well outside the SN. We then demonstrated that the reduction in weight gain following excessive-doses can be dissociated from NRTN in the targeted SN, and is linked to mistargeted NRTN in the diencephalon. We also showed that prior destruction of the dopaminergic SN neurons via 6-OHDA had no impact on the weight loss phenomenon, further dissociating neurotrophic exposure to the SN as the culprit for weight changes. Finally, low AAV2-NRTN doses provided significant neuroprotection against 6-OHDA toxicity, establishing a wide therapeutic index for nigral targeting. These data support targeting the SN with AAV2-NRTN in PD patients, demonstrating that properly targeted and scaled AAV2-NRTN provides safe and effective NRTN expression. They also provided the means to define an appropriate human-equivalent dose for proceeding into an ongoing clinical trial, using empirically-based scaling to account for marked differences in SN volume between species.
Collapse
|
30
|
Harada K, Horiuchi H, Tanabe K, Carter RG, Esumi T, Kubo M, Hioki H, Fukuyama Y. Asymmetric synthesis of (−)-chicanine using a highly regioselective intramolecular Mitsunobu reaction and revision of its absolute configuration. Tetrahedron Lett 2011. [DOI: 10.1016/j.tetlet.2011.03.154] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
31
|
Recent progress in cell therapy for basal ganglia disorders with emphasis on menstrual blood transplantation in stroke. Neurosci Biobehav Rev 2011; 36:177-90. [PMID: 21645544 DOI: 10.1016/j.neubiorev.2011.05.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 04/25/2011] [Accepted: 05/20/2011] [Indexed: 12/13/2022]
Abstract
Cerebrovascular diseases are the third leading cause of death and the primary cause of long-term disability in the United States. The only approved therapy for stroke is tPA, strongly limited by the short therapeutic window and hemorrhagic complications, therefore excluding most patients from its benefits. Parkinson's and Huntington's disease are the other two most studied basal ganglia diseases and, as stroke, have very limited treatment options. Inflammation is a key feature in central nervous system disorders and it plays a dual role, either improving injury in early phases or impairing neural survival at later stages. Stem cells can be opportunely used to modulate inflammation, abrogate cell death and, therefore, preserve neural function. We here discuss the role of stem cells as restorative treatments for basal ganglia disorders, including Parkinson's disease, Huntington's disease and stroke, with special emphasis to the recently investigated menstrual blood stem cells. We highlight the availability, proliferative capacity, pluripotentiality and angiogenic features of these cells and explore their present and future experimental and clinical applications.
Collapse
|
32
|
Xu J, Trzoss L, Chang WK, Theodorakis EA. Enantioselective total synthesis of (-)-jiadifenolide. Angew Chem Int Ed Engl 2011; 50:3672-6. [PMID: 21400650 PMCID: PMC3159889 DOI: 10.1002/anie.201100313] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Indexed: 11/07/2022]
Abstract
The first total synthesis of jiadifenolide (1 ), a potent neurotrophic modulator, has been reported. Highlights of the synthesis include: construction of the B ring via an asymmetric Robinson annulation; assembly of the E ring lactone via a novel acid-induced cascade reaction; and Pd(0)-mediated carbomethoxylation and methylation reactions for the construction of the C and A rings respectively.
Collapse
Affiliation(s)
- Jing Xu
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California, 92093-0358, USA
| | - Lynnie Trzoss
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California, 92093-0358, USA
| | - Weng K. Chang
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California, 92093-0358, USA
| | - Emmanuel. A. Theodorakis
- Department of Chemistry and Biochemistry, University of California, San Diego, 9500 Gilman Drive, La Jolla, California, 92093-0358, USA
| |
Collapse
|
33
|
Xu J, Trzoss L, Chang WK, Theodorakis EA. Enantioselective Total Synthesis of (−)-Jiadifenolide. Angew Chem Int Ed Engl 2011. [DOI: 10.1002/ange.201100313] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
34
|
Sapkota K, Kim S, Park SE, Kim SJ. Detoxified extract of Rhus verniciflua stokes inhibits rotenone-induced apoptosis in human dopaminergic cells, SH-SY5Y. Cell Mol Neurobiol 2011; 31:213-23. [PMID: 21061154 PMCID: PMC11498594 DOI: 10.1007/s10571-010-9609-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Accepted: 09/28/2010] [Indexed: 12/12/2022]
Abstract
Rhus verniciflua Stokes (RVS), traditionally used as a food supplement and in traditional herbal medicine for centuries in Korea, is known to possess various pharmacological properties. Environmental neurotoxins such as rotenone, a specific inhibitor of complex I provide models of Parkinson's disease (PD) both in vivo and in vitro. In this study, we investigated the neuroprotective effect of RVS against rotenone-induced toxicity in human dopaminergic cells, SH-SY5Y. Cells exposed to rotenone for 24 h-induced cellular injury and apoptotic cell death. Pretreatment of cells with RVS provided significant protection to SH-SY5Y cells. Further, RVS offered remarkable protection against rotenone-induced oxidative stress and markedly inhibited mitochondrial membrane potential (MMP) disruption. RVS also attenuated the up-regulation of Bax, Caspase-9 and Caspase-3 and down-regulation of Bcl-2. Moreover, pretreatment with RVS prevented the decrease in tyrosine hydroxylase (TH) levels in SH-SY5Y cells. Interestingly, RVS conferred profound protection to human dopaminergic cells by preventing the downregulation of brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF). These results suggest that RVS may protect dopaminergic neurons against rotenone-induced apoptosis by multiple functions and contribute to neuroprotection in neurodegenerative diseases, such as PD.
Collapse
Affiliation(s)
- Kumar Sapkota
- Department of Biotechnology, Chosun University, 375 Seosuk-dong, Dong-gu, Gwang-ju, 501-759 Republic of Korea
- Central Department of Zoology, Tribhuvan University, Kirtipur, Kathmandu,
Nepal
| | - Seung Kim
- Department of Alternative medicine, Gwangju University, Gwangju, 503-703 Republic of Korea
| | - Se-Eun Park
- Department of Biotechnology, Chosun University, 375 Seosuk-dong, Dong-gu, Gwang-ju, 501-759 Republic of Korea
| | - Sung-Jun Kim
- Department of Biotechnology, Chosun University, 375 Seosuk-dong, Dong-gu, Gwang-ju, 501-759 Republic of Korea
| |
Collapse
|
35
|
Baratchi S, Kanwar RK, Kanwar JR. Novel survivin mutant protects differentiated SK-N-SH human neuroblastoma cells from activated T-cell neurotoxicity. J Neuroimmunol 2010; 233:18-28. [PMID: 21129784 DOI: 10.1016/j.jneuroim.2010.10.036] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Revised: 10/27/2010] [Accepted: 10/29/2010] [Indexed: 01/08/2023]
Abstract
Currently, there are no known treatments for protection of axonal loss associated with neuroinflammatory diseases such as multiple sclerosis (MS). Survivin is a member of the inhibitors of the apoptosis (IAP) family of proteins that its neuroprotective effects have not been studied. We demonstrate here that SurR9-C84A, a survivin mutant, exhibits a neuroprotective role against the cytotoxic effects of activated T-cell infiltrates, such as granzyme B (GrB). The activated T-cell supernatants induce toxicity on differentiated SK-N-SH cells, which is associated with the loss of Ca(2+) homeostasis, the increased population of dead cells, mitochondrial membrane depolarisation, and the accelerated expression of cyclinD1, caspase3 and Fas, as observed for most apoptotic cells. Alternatively, the pre-treatment with SurR9-C84A reduces the population of dead cells by balancing the cytosolic Ca(2+) homeostasis, decreasing the level of mitochondrial depolarisation, and also reducing the expression of cyclinD1 and caspase3. Our findings suggest that SurR9-C84A has a neuroprotective effect against the cytotoxins existing in activated T-cell supernatants including GrB.
Collapse
Affiliation(s)
- Sara Baratchi
- Laboratory of Immunology and Molecular Biomedical Research, Centre for Biotechnology and Interdisciplinary Biosciences (BioDeakin), Institute for Technology Research and Innovation (ITRI), Deakin University, Waurn Ponds, Victoria 3217, Australia
| | | | | |
Collapse
|
36
|
Altman RA, Nilsson BL, Overman LE, de Alaniz JR, Rohde JM, Taupin V. Total synthesis of (+)-nankakurines A and B and (±)-5-epi-nankakurine A. J Org Chem 2010; 75:7519-34. [PMID: 20958075 PMCID: PMC3038189 DOI: 10.1021/jo101619d] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The first total syntheses of the Lycopodium alkaloids (+)-nankakurine A (2), (+)-nankakurine B (3), and the originally purported structure 1 of nankakurine A were accomplished. The syntheses of 2 and 3 feature a demanding intramolecular azomethine imine cycloaddition as the key step for generating the octahydro-3,5-ethanoquinoline moiety and installing the correct relative configuration at the spiropiperidine ring juncture. The cyclization precursor was prepared from octahydronaphthalene ketone 50, which was assembled from enone (+)-9 and diene 48 by a cationic Diels-Alder reaction. The Diels-Alder reactants were synthesized from 5-hexyn-1-ol (16) and (+)-pulegone (49), respectively. The tetracyclic ring system of 1 was generated using an unprecedented nitrogen-terminated aza-Prins cyclization cascade. The enantioselective total syntheses of (+)-nankakurine A (2) and (+)-nankakurine B (3) establish the relative and absolute configuration of these alkaloids and are sufficiently concise that substantial quantities of 2 and 3 were prepared for biological studies. (+)-Nankakurine A and (+)-nankakurine B showed no effect on neurite outgrowth in rat hippocampal H-19 cells over a concentration range of 0.3-10 μM.
Collapse
Affiliation(s)
- Ryan A. Altman
- Department of Chemistry, 1102 Natural Sciences II, University of California, Irvine, Irvine, CA 92697-2025
| | - Bradley L. Nilsson
- Department of Chemistry, 1102 Natural Sciences II, University of California, Irvine, Irvine, CA 92697-2025
| | - Larry E. Overman
- Department of Chemistry, 1102 Natural Sciences II, University of California, Irvine, Irvine, CA 92697-2025
| | - Javier Read de Alaniz
- Department of Chemistry, 1102 Natural Sciences II, University of California, Irvine, Irvine, CA 92697-2025
| | - Jason M. Rohde
- Department of Chemistry, 1102 Natural Sciences II, University of California, Irvine, Irvine, CA 92697-2025
| | - Veronique Taupin
- Aging Department, sanofi-aventis R&D, 1 Avenue Pierre Brossolette, 91385 Chilly-Mazarin, France
| |
Collapse
|
37
|
Arien-Zakay H, Lecht S, Nagler A, Lazarovici P. Human umbilical cord blood stem cells: rational for use as a neuroprotectant in ischemic brain disease. Int J Mol Sci 2010; 11:3513-28. [PMID: 20957109 PMCID: PMC2956109 DOI: 10.3390/ijms11093513] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 09/15/2010] [Accepted: 09/15/2010] [Indexed: 01/19/2023] Open
Abstract
The use of stem cells for reparative medicine was first proposed more than three decades ago. Hematopoietic stem cells from bone marrow, peripheral blood and human umbilical cord blood (CB) have gained major use for treatment of hematological indications. CB, however, is also a source of cells capable of differentiating into various non-hematopoietic cell types, including neural cells. Several animal model reports have shown that CB cells may be used for treatment of neurological injuries. This review summarizes the information available on the origin of CB-derived neuronal cells and the mechanisms proposed to explain their action. The potential use of stem/progenitor cells for treatment of ischemic brain injuries is discussed. Issues that remain to be resolved at the present stage of preclinical trials are addressed.
Collapse
Affiliation(s)
- Hadar Arien-Zakay
- The School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel; E-Mails: (H.A.-Z.); (S.L.)
- Division of Hematology and Cord Blood Bank, Chaim Sheba Medical Center, Tel-Hashomer, Israel; E-Mail: (A.N.)
| | - Shimon Lecht
- The School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel; E-Mails: (H.A.-Z.); (S.L.)
| | - Arnon Nagler
- Division of Hematology and Cord Blood Bank, Chaim Sheba Medical Center, Tel-Hashomer, Israel; E-Mail: (A.N.)
| | - Philip Lazarovici
- The School of Pharmacy Institute for Drug Research, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel; E-Mails: (H.A.-Z.); (S.L.)
- * Author to whom correspondence should be addressed: E-Mail: ; Tel.: 972-2-6758-729; Fax: 972-2-6757-490
| |
Collapse
|
38
|
Nai Q, Wang X, Jin Y, Sun D, Li M, Hu B, Zhang X. Ciliary neurotrophic factor enhances nicotinic synaptic transmission in sympathetic neurons. J Neurosci Res 2010; 88:887-95. [PMID: 19830843 DOI: 10.1002/jnr.22260] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Nicotinic acetylcholine receptors mediate fast synaptic transmission in both central and peripheral nervous systems. These receptors play important roles in various physiological functions and are involved in different neurological diseases. A disruption in nicotinic receptor-mediated synaptic transmission due to the loss of nAChRs was detected in the brains of patients with Parkinson's disease and Alzheimer's disease. Although ciliary neurotrophic factor (CNTF) has been reported to promote the cholinergic properties by increasing the production and storage of acetylcholine, it is still unclear whether CNTF can enhance nicotinic synaptic neurotransmission. In this study, we found that CNTF dramatically enhanced the frequency and amplitude of nicotinic excitatory post-synaptic currents in rat superior cervical ganglion neurons maintained in a medium supplemented with nerve growth factor. Moreover, the number of neurons displaying nicotinic synaptic currents was also significantly increased by CNTF. These results suggest that CNTF could enhance nicotinic synaptic transmission via both presynaptic and postsynaptic mechanisms. The findings of this study reinforce the rationale for the usage of combinations of different neurotrophic factors for the therapy of neurodegenerative diseases.
Collapse
Affiliation(s)
- Qiang Nai
- Department of Anatomy and Neurobiology, University of Tennessee, Health Science Center, Memphis, Tennessee, USA.
| | | | | | | | | | | | | |
Collapse
|
39
|
Cheng X, Waters SP. Concise total syntheses of the Lycopodium alkaloids (+/-)-nankakurines A and B via luciduline. Org Lett 2010; 12:205-7. [PMID: 20014779 PMCID: PMC2804776 DOI: 10.1021/ol902455y] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Total syntheses of the Lycopodium alkaloids nankakurines A and B have been accomplished in 6 and 7 steps, respectively, via a sequence that passes through a third Lycopodium alkaloid, luciduline, and forgoes the use of protecting groups on nitrogen. Key features include a short preparation of luciduline followed by a concise and stereoselective aminoallylation/ring-closing metathesis protocol to fashion the spiropiperidine ring common to nankakurines A and B.
Collapse
Affiliation(s)
- Xiayun Cheng
- Department of Chemistry, The University of Vermont, 82 University Place, Burlington, Vermont 05405
| | - Stephen P. Waters
- Department of Chemistry, The University of Vermont, 82 University Place, Burlington, Vermont 05405
| |
Collapse
|
40
|
|
41
|
Jessen HJ, Barbaras D, Hamburger M, Gademann K. Total Synthesis and Neuritotrophic Activity of Farinosone C and Derivatives. Org Lett 2009; 11:3446-9. [DOI: 10.1021/ol901277q] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Henning J. Jessen
- Swiss Federal Institute of Technology (EPFL), Chemical Synthesis Laboratory (SB-ISIC-LSYNC), 1015 Lausanne, Switzerland, Swiss Federal Institute of Technology (ETH), Laboratorium für Organische Chemie, 8093 Zürich, Switzerland, and University of Basel, Institute of Pharmaceutical Biology, 4056 Basel, Switzerland
| | - Damien Barbaras
- Swiss Federal Institute of Technology (EPFL), Chemical Synthesis Laboratory (SB-ISIC-LSYNC), 1015 Lausanne, Switzerland, Swiss Federal Institute of Technology (ETH), Laboratorium für Organische Chemie, 8093 Zürich, Switzerland, and University of Basel, Institute of Pharmaceutical Biology, 4056 Basel, Switzerland
| | - Matthias Hamburger
- Swiss Federal Institute of Technology (EPFL), Chemical Synthesis Laboratory (SB-ISIC-LSYNC), 1015 Lausanne, Switzerland, Swiss Federal Institute of Technology (ETH), Laboratorium für Organische Chemie, 8093 Zürich, Switzerland, and University of Basel, Institute of Pharmaceutical Biology, 4056 Basel, Switzerland
| | - Karl Gademann
- Swiss Federal Institute of Technology (EPFL), Chemical Synthesis Laboratory (SB-ISIC-LSYNC), 1015 Lausanne, Switzerland, Swiss Federal Institute of Technology (ETH), Laboratorium für Organische Chemie, 8093 Zürich, Switzerland, and University of Basel, Institute of Pharmaceutical Biology, 4056 Basel, Switzerland
| |
Collapse
|
42
|
Nilsson BL, Overman LE, de Alaniz JR, Rohde JM. Enantioselective total syntheses of nankakurines A and B: confirmation of structure and establishment of absolute configuration. J Am Chem Soc 2008; 130:11297-9. [PMID: 18680252 PMCID: PMC3074941 DOI: 10.1021/ja804624u] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Total syntheses of (+)-nankakurine A (2) and (+)-nankakurine B (3) were accomplished by a sequence that employs an intramolecular dipolar cycloaddition of an azomethine imine intermediate to form the azatricyclic moiety and establish the relative configuration of the spiropiperidine ring. These syntheses, together with the synthesis of the originally purported structure 1 of nankakurine A, rigorously establish the relative and absolute configuration of these structurally unusual Lycopodium alkaloids. The syntheses are sufficiently concise that gram quantities of (+)-nankakurine A (2) and (+)-nankakurine B (3) will be available for further biological studies.
Collapse
Affiliation(s)
| | - Larry E. Overman
- Department of Chemistry, University of California, Irvine, 1102 Natural Sciences II, Irvine, California 92697-2025
| | - Javier Read de Alaniz
- Department of Chemistry, University of California, Irvine, 1102 Natural Sciences II, Irvine, California 92697-2025
| | | |
Collapse
|
43
|
Pardridge WM. Re-Engineering Biopharmaceuticals for Delivery to Brain with Molecular Trojan Horses. Bioconjug Chem 2008; 19:1327-38. [DOI: 10.1021/bc800148t] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- William M. Pardridge
- Department of Medicine, University of California at Los Angeles, Los Angeles, California 90024
| |
Collapse
|
44
|
Boado RJ, Zhang Y, Zhang Y, Pardridge WM. Genetic engineering, expression, and activity of a fusion protein of a human neurotrophin and a molecular Trojan horse for delivery across the human blood-brain barrier. Biotechnol Bioeng 2007; 97:1376-86. [PMID: 17286273 DOI: 10.1002/bit.21369] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Neurotrophins, such as brain derived neurotrophic factor (BDNF), do not cross the blood-brain barrier (BBB). Certain monoclonal antibodies (MAb) to the human insulin receptor (HIR) do cross the BBB via receptor-mediated transport, and can act as a molecular Trojan horse to ferry across the BBB an attached drug. A genetically engineered fusion protein was produced whereby the amino terminus of human BDNF is fused to the carboxyl terminus of the heavy chain of a chimeric HIRMAb. The HIRMAb-BDNF fusion protein reacted equally with antibodies to human IgG and BDNF. The bi-functionality of the fusion protein was retained as the affinity of the fusion protein for the HIR was identical to that of the chimeric HIRMAb, and the affinity of the fusion protein for the trkB receptor was identical to that of BDNF. The fusion protein was equi-potent with BDNF in a neuroprotection assay in human neural cells. The pharmacokinetics (PK) of the fusion protein was examined in the adult Rhesus monkey. The mean residence time (MRT) of the fusion protein in blood was >100-fold longer than the MRT of BDNF. Therapeutic levels of BDNF were produced in primate brain following the intravenous administration of the fusion protein. A fusion protein tandem vector was engineered that allowed for isolation of a CHO cell line that produced the fusion protein at high levels in serum free medium. Neurotrophins, such as BDNF, can be re-formulated to enable these molecules to cross the human BBB, and such fusion proteins represent a new class of human neurotherapeutics.
Collapse
Affiliation(s)
- Ruben J Boado
- ArmaGen Technologies Inc., Santa Monica, California, USA
| | | | | | | |
Collapse
|
45
|
Affiliation(s)
- Goverdhan Mehta
- Department of Organic Chemistry, Indian Institute of Science, Bangalore.
| | | |
Collapse
|
46
|
Niewiadomska G, Baksalerska-Pazera M, Gasiorowska A, Mietelska A. Nerve Growth Factor Differentially Affects Spatial and Recognition Memory in Aged Rats. Neurochem Res 2006; 31:1481-90. [PMID: 17111224 DOI: 10.1007/s11064-006-9209-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2006] [Accepted: 10/20/2006] [Indexed: 10/23/2022]
Abstract
In rats, object discrimination depends on the integrity of the cholinergic system, thus it could be expected that nerve growth factor (NGF) can improve the behavior in aged subjects. The interactive effect of age and cholinergic improvement was assessed behaviorally in young and aged rats. Animals were injected by infusion of NGF into the lateral ventricles and they were tested in two behavioral tasks: an object-location and an object-recognition task. Spatial and recognition memory were assessed in an open field containing five different objects. Rats were submitted to six consecutive sessions. Both age-groups showed comparable habituation of exploratory response in Session 1-4. Discrimination index (DI) was calculated to assess responses to spatial change in Session 5 and object change in Session 6. Control young and aged rats were able to discriminate between familiar and novel object, however DI was lower in aged rats. Treatment with NGF induced decline of object discrimination in both age-groups. Different results were obtained in spatial displacement test. NGF was able to improve spatial memory in aged rats, but had no effect in young controls. These data confer on NGF potential role in improving spatial but not episodic memory in aged rats.
Collapse
Affiliation(s)
- G Niewiadomska
- Department of Neurophysiology, Nencki Institute for Experimental Biology, 02-093, Warsaw, Poland.
| | | | | | | |
Collapse
|
47
|
Tesseur I, Zou K, Esposito L, Bard F, Berber E, Can JV, Lin AH, Crews L, Tremblay P, Mathews P, Mucke L, Masliah E, Wyss-Coray T. Deficiency in neuronal TGF-beta signaling promotes neurodegeneration and Alzheimer's pathology. J Clin Invest 2006; 116:3060-9. [PMID: 17080199 PMCID: PMC1626127 DOI: 10.1172/jci27341] [Citation(s) in RCA: 281] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2005] [Accepted: 08/01/2006] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by progressive neurodegeneration and cerebral accumulation of the beta-amyloid peptide (Abeta), but it is unknown what makes neurons susceptible to degeneration. We report that the TGF-beta type II receptor (TbetaRII) is mainly expressed by neurons, and that TbetaRII levels are reduced in human AD brain and correlate with pathological hallmarks of the disease. Reducing neuronal TGF-beta signaling in mice resulted in age-dependent neurodegeneration and promoted Abeta accumulation and dendritic loss in a mouse model of AD. In cultured cells, reduced TGF-beta signaling caused neuronal degeneration and resulted in increased levels of secreted Abeta and beta-secretase-cleaved soluble amyloid precursor protein. These results show that reduced neuronal TGF-beta signaling increases age-dependent neurodegeneration and AD-like disease in vivo. Increasing neuronal TGF-beta signaling may thus reduce neurodegeneration and be beneficial in AD.
Collapse
Affiliation(s)
- Ina Tesseur
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.
Gladstone Institute of Neurological Disease and
Department of Neurology, University of California San Francisco, San Francisco, California, USA.
Elan Pharmaceuticals Inc., South San Francisco, California, USA.
Departments of Neuroscience and Pathology, University of California San Diego, San Diego, California, USA.
Center for Dementia Research, New York University School of Medicine, Orangeburg, New York, USA.
Geriatric Research, Education, and Clinical Center (GRECC), VA Palo Alto Health Care System, Palo Alto, California, USA
| | - Kun Zou
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.
Gladstone Institute of Neurological Disease and
Department of Neurology, University of California San Francisco, San Francisco, California, USA.
Elan Pharmaceuticals Inc., South San Francisco, California, USA.
Departments of Neuroscience and Pathology, University of California San Diego, San Diego, California, USA.
Center for Dementia Research, New York University School of Medicine, Orangeburg, New York, USA.
Geriatric Research, Education, and Clinical Center (GRECC), VA Palo Alto Health Care System, Palo Alto, California, USA
| | - Luke Esposito
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.
Gladstone Institute of Neurological Disease and
Department of Neurology, University of California San Francisco, San Francisco, California, USA.
Elan Pharmaceuticals Inc., South San Francisco, California, USA.
Departments of Neuroscience and Pathology, University of California San Diego, San Diego, California, USA.
Center for Dementia Research, New York University School of Medicine, Orangeburg, New York, USA.
Geriatric Research, Education, and Clinical Center (GRECC), VA Palo Alto Health Care System, Palo Alto, California, USA
| | - Frederique Bard
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.
Gladstone Institute of Neurological Disease and
Department of Neurology, University of California San Francisco, San Francisco, California, USA.
Elan Pharmaceuticals Inc., South San Francisco, California, USA.
Departments of Neuroscience and Pathology, University of California San Diego, San Diego, California, USA.
Center for Dementia Research, New York University School of Medicine, Orangeburg, New York, USA.
Geriatric Research, Education, and Clinical Center (GRECC), VA Palo Alto Health Care System, Palo Alto, California, USA
| | - Elisabeth Berber
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.
Gladstone Institute of Neurological Disease and
Department of Neurology, University of California San Francisco, San Francisco, California, USA.
Elan Pharmaceuticals Inc., South San Francisco, California, USA.
Departments of Neuroscience and Pathology, University of California San Diego, San Diego, California, USA.
Center for Dementia Research, New York University School of Medicine, Orangeburg, New York, USA.
Geriatric Research, Education, and Clinical Center (GRECC), VA Palo Alto Health Care System, Palo Alto, California, USA
| | - Judith Van Can
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.
Gladstone Institute of Neurological Disease and
Department of Neurology, University of California San Francisco, San Francisco, California, USA.
Elan Pharmaceuticals Inc., South San Francisco, California, USA.
Departments of Neuroscience and Pathology, University of California San Diego, San Diego, California, USA.
Center for Dementia Research, New York University School of Medicine, Orangeburg, New York, USA.
Geriatric Research, Education, and Clinical Center (GRECC), VA Palo Alto Health Care System, Palo Alto, California, USA
| | - Amy H. Lin
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.
Gladstone Institute of Neurological Disease and
Department of Neurology, University of California San Francisco, San Francisco, California, USA.
Elan Pharmaceuticals Inc., South San Francisco, California, USA.
Departments of Neuroscience and Pathology, University of California San Diego, San Diego, California, USA.
Center for Dementia Research, New York University School of Medicine, Orangeburg, New York, USA.
Geriatric Research, Education, and Clinical Center (GRECC), VA Palo Alto Health Care System, Palo Alto, California, USA
| | - Leslie Crews
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.
Gladstone Institute of Neurological Disease and
Department of Neurology, University of California San Francisco, San Francisco, California, USA.
Elan Pharmaceuticals Inc., South San Francisco, California, USA.
Departments of Neuroscience and Pathology, University of California San Diego, San Diego, California, USA.
Center for Dementia Research, New York University School of Medicine, Orangeburg, New York, USA.
Geriatric Research, Education, and Clinical Center (GRECC), VA Palo Alto Health Care System, Palo Alto, California, USA
| | - Patrick Tremblay
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.
Gladstone Institute of Neurological Disease and
Department of Neurology, University of California San Francisco, San Francisco, California, USA.
Elan Pharmaceuticals Inc., South San Francisco, California, USA.
Departments of Neuroscience and Pathology, University of California San Diego, San Diego, California, USA.
Center for Dementia Research, New York University School of Medicine, Orangeburg, New York, USA.
Geriatric Research, Education, and Clinical Center (GRECC), VA Palo Alto Health Care System, Palo Alto, California, USA
| | - Paul Mathews
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.
Gladstone Institute of Neurological Disease and
Department of Neurology, University of California San Francisco, San Francisco, California, USA.
Elan Pharmaceuticals Inc., South San Francisco, California, USA.
Departments of Neuroscience and Pathology, University of California San Diego, San Diego, California, USA.
Center for Dementia Research, New York University School of Medicine, Orangeburg, New York, USA.
Geriatric Research, Education, and Clinical Center (GRECC), VA Palo Alto Health Care System, Palo Alto, California, USA
| | - Lennart Mucke
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.
Gladstone Institute of Neurological Disease and
Department of Neurology, University of California San Francisco, San Francisco, California, USA.
Elan Pharmaceuticals Inc., South San Francisco, California, USA.
Departments of Neuroscience and Pathology, University of California San Diego, San Diego, California, USA.
Center for Dementia Research, New York University School of Medicine, Orangeburg, New York, USA.
Geriatric Research, Education, and Clinical Center (GRECC), VA Palo Alto Health Care System, Palo Alto, California, USA
| | - Eliezer Masliah
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.
Gladstone Institute of Neurological Disease and
Department of Neurology, University of California San Francisco, San Francisco, California, USA.
Elan Pharmaceuticals Inc., South San Francisco, California, USA.
Departments of Neuroscience and Pathology, University of California San Diego, San Diego, California, USA.
Center for Dementia Research, New York University School of Medicine, Orangeburg, New York, USA.
Geriatric Research, Education, and Clinical Center (GRECC), VA Palo Alto Health Care System, Palo Alto, California, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA.
Gladstone Institute of Neurological Disease and
Department of Neurology, University of California San Francisco, San Francisco, California, USA.
Elan Pharmaceuticals Inc., South San Francisco, California, USA.
Departments of Neuroscience and Pathology, University of California San Diego, San Diego, California, USA.
Center for Dementia Research, New York University School of Medicine, Orangeburg, New York, USA.
Geriatric Research, Education, and Clinical Center (GRECC), VA Palo Alto Health Care System, Palo Alto, California, USA
| |
Collapse
|
48
|
Simonato M, Tongiorgi E, Kokaia M. Angels and demons: neurotrophic factors and epilepsy. Trends Pharmacol Sci 2006; 27:631-8. [PMID: 17055067 DOI: 10.1016/j.tips.2006.10.002] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2006] [Revised: 09/11/2006] [Accepted: 10/09/2006] [Indexed: 12/29/2022]
Abstract
Several lines of evidence indicate that neurotrophic factors (NTFs) could be key causal mediators in the development of acquired epileptic syndromes. Yet the trophic properties of NTFs indicate that they might be used to treat epilepsy-associated damage. Accordingly, different NTFs, or even the same NTF, could produce functionally contrasting effects in the context of epilepsy. Recent experimental evidence begins to shed light on the mechanisms underlying these contrasting effects. Understanding these mechanisms will be instrumental for the development of effective therapies, which must be based on a careful consideration of the biological properties of NTFs. Here, we critically evaluate new information emerging in this area and discuss its implications for clinical treatment.
Collapse
Affiliation(s)
- Michele Simonato
- Department of Clinical and Experimental Medicine, Section of Pharmacology, University of Ferrara, Ferrara 44100, Italy.
| | | | | |
Collapse
|
49
|
Howe CL, Mayoral S, Rodriguez M. Activated microglia stimulate transcriptional changes in primary oligodendrocytes via IL-1beta. Neurobiol Dis 2006; 23:731-9. [PMID: 16887357 DOI: 10.1016/j.nbd.2006.06.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2006] [Revised: 06/17/2006] [Accepted: 06/22/2006] [Indexed: 10/24/2022] Open
Abstract
No therapy currently exists to repair demyelinated lesions in multiple sclerosis. However, the use of IgM antibodies may provide a valuable therapeutic avenue for evoking such repair. Unfortunately, the mechanism of immunoglobulin action in CNS repair is currently unknown but may depend upon complex interactions between multiple cell types rather than upon direct activation of a single cell type. Using rat mixed glial cultures containing oligodendrocytes, microglia, and astrocytes, we found that the Fc portion of human IgM shifts microglia to an activated phenotype, reduces glial proliferation, upregulates a variety of immediate early genes, including JunB, Egr-1, and c-Fos, and stimulates microglial production and release of IL-1beta. Microglia-derived IL-1beta consequently triggers transcriptional upregulation of immediate early genes such as c-Jun, Egr-1, and c-Fos in the mixed glial cultures, and stimulates the upregulation of late response genes such as lipocalin in purified oligodendrocytes. Treatment with an IL-1beta receptor antagonist abrogates the effects of Fcmu on glial proliferation and prevents the upregulation of lipocalin in response to Fcmu, but does not prevent Fcmu-mediated upregulation of IL-1beta, suggesting that IL-1beta mediates at least some of the downstream effects of Fcmu in mixed glial cultures. We hypothesize that Fcmu-stimulated IL-1beta-induced upregulation of immediate early and late response genes in oligodendrocytes may promote CNS repair.
Collapse
Affiliation(s)
- Charles L Howe
- Department of Neuroscience, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| | | | | |
Collapse
|
50
|
Wilson RM, Danishefsky SJ. Applications of total synthesis to problems in neurodegeneration: Fascinating chemistry along the way. Acc Chem Res 2006; 39:539-49. [PMID: 16906750 DOI: 10.1021/ar068018n] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The possibility for the application of organic synthesis to the discovery of new agents in combating neurodegenerative disorders is described. Our focus has been on agents derived from natural-product leads and natural products themselves prepared through total synthesis. Herein, we describe some of the chemistry as well as interesting observations made along the way.
Collapse
Affiliation(s)
- Rebecca M Wilson
- Laboratory for Bioorganic Chemistry, Sloan-Kettering Institute for Cancer Research, 1275 York Avenue, New York, New York 10021, USA
| | | |
Collapse
|