1
|
Yao S, Zhang W, Xiao J, Zhang Z, Wang L, Ai H, Wu X, Chen A, Zhuang X. Simultaneous determination of HD56, a novel prodrug, and its active metabolite in cynomolgus monkey plasma using LC-MS/MS for elucidating its pharmacokinetic profile. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1235:124045. [PMID: 38367406 DOI: 10.1016/j.jchromb.2024.124045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/31/2023] [Revised: 01/25/2024] [Accepted: 02/07/2024] [Indexed: 02/19/2024]
Abstract
An LC-MS/MS method was developed and validated for the simultaneous determination of the carboxylic acid ester precursor HD56 and the active product HD561 in cynomolgus monkey plasma. Then, the pharmacokinetic characteristics of both compounds following single and multiple i.g. administrations in cynomolgus monkeys were elucidated. In the method, chromatographic separation was achieved with a C18 reversed-phase column and the target quantification was carried out by an electrospray ionization (ESI) source coupled with triple quadrupole mess detector in positive ionization mode with multiple reaction monitoring (MRM) approach. Using the quantification method, the in vitro stability of HD56 in plasma and HD56 pharmacokinetic behavior after i.g. administration in cynomolgus monkey were investigated. It was approved that HD56 did convert into HD561 post-administration. The overall systemic exposure of HD561 post-conversion from HD56 accounted for only about 17% of HD56. After repeated administration at the same dose, there was no significant difference in exposure levels of both HD56 and HD561. However, after multiple dosing, the exposure of HD56 tended to decrease while that of HD561 tended to increase, resulting in a 30% in the exposure ratio. Remarkably, with a carboxylesterase (CES) activity profile akin to humans, the observed in vivo pharmacokinetic profile in cynomolgus monkeys holds promise for predicting HD56/HD561 PK profiles in humans.
Collapse
Affiliation(s)
- Shi Yao
- School of Chemical and Pharmaceutical Engineering Hebei University of Science and Technology Shijiazhuang 050018 China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Wenpeng Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Junhai Xiao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Zhiwei Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Lingchao Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Hengxiao Ai
- School of Chemical and Pharmaceutical Engineering Hebei University of Science and Technology Shijiazhuang 050018 China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Xia Wu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Aibing Chen
- School of Chemical and Pharmaceutical Engineering Hebei University of Science and Technology Shijiazhuang 050018 China.
| | - Xiaomei Zhuang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| |
Collapse
|
2
|
Rasool A, Manzoor R, Ullah K, Afzal R, Ul-Haq A, Imran H, Kaleem I, Akhtar T, Farrukh A, Hameed S, Bashir S. Oxidative Stress and Dopaminergic Metabolism: A Major PD Pathogenic Mechanism and Basis of Potential Antioxidant Therapies. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:852-864. [PMID: 37303175 DOI: 10.2174/1871527322666230609141519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 08/08/2022] [Revised: 02/19/2023] [Accepted: 03/14/2023] [Indexed: 06/13/2023]
Abstract
Reactive oxygen species (ROS)-induced oxidative stress triggers the vicious cycle leading to the degeneration of dopaminergic neurons in the nigra pars compacta. ROS produced during the metabolism of dopamine is immediately neutralized by the endogenous antioxidant defense system (EADS) under physiological conditions. Aging decreases the vigilance of EADS and makes the dopaminergic neurons more vulnerable to oxidative stress. As a result, ROS left over by EADS oxidize the dopamine-derived catechols and produces a number of reactive dopamine quinones, which are precursors to endogenous neurotoxins. In addition, ROS causes lipid peroxidation, uncoupling of the electron transport chain, and DNA damage, which lead to mitochondrial dysfunction, lysosomal dysfunction, and synaptic dysfunction. The mutations in genes such as DNAJC6, SYNJ1, SH3GL2, LRRK2, PRKN, and VPS35 caused by ROS have been associated with synaptic dysfunction and the pathogenesis of Parkinson's disease (PD). The available drugs that are used against PD can only delay the progression of the disease, but they produce various side effects. Through their antioxidant activity, flavonoids can substantiate the EADS of dopaminergic neurons and disrupt the vicious cycle incepted by oxidative stress. In this review, we show how the oxidative metabolism of dopamine generates ROS and dopamine-quinones, which then exert unrestrained OS, causing mutations in several genes involved in the proper functioning of mitochondrion, synapse, and lysosome. Besides, we also present some examples of approved drugs used for the treatment of PD, therapies in the clinical trial phase, and an update on the flavonoids that have been tested to boost the EADS of dopaminergic neurons.
Collapse
Affiliation(s)
- Aamir Rasool
- Institute for Synthetic Biosystem, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing 100081, PR China
- Institute of Biochemistry, University of Balochistan, Quetta 87300, Pakistan
| | - Robina Manzoor
- Beijing Key Laboratory for Separation and Analysis in Biomedicine and Pharmaceuticals, School of Life Science, Beijing Institute of Technology, Beijing 100081, PR China
- Faculty of Marine Sciences, Lasbella University of Agriculture Water and Marine Sciences, Uthal 90050, Pakistan
| | - Kaleem Ullah
- Department of Microbiology, University of Balochistan, Quetta 87300, Pakistan
| | - Ramsha Afzal
- Department of Brain Science, Ajou University School of Medicine, Suwon 16499, Republic of Korea
| | - Asad Ul-Haq
- Division of Rheumatology, Department of Internal Medicine, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Hadia Imran
- Department of Biosciences, COMSATS University Islamabad, Pakistan
| | - Imdad Kaleem
- Department of Biosciences, COMSATS University Islamabad, Pakistan
| | | | - Anum Farrukh
- Department of General Medicine, Fauji Foundation Hospital (FFH), Rawalpindi, Pakistan
| | - Sahir Hameed
- National Institute for Genomics and Advanced Biotechnology (N.I.G.A.B.) National Agriculture Research Centre Islamabad, Pakistan
| | - Shahid Bashir
- Neurosciences Center, King Fahad Specialist Hospital Dammam, P.O. Box 15215, Dammam 31444, Saudi Arabia
| |
Collapse
|
3
|
Henrich MT, Oertel WH, Surmeier DJ, Geibl FF. Mitochondrial dysfunction in Parkinson's disease - a key disease hallmark with therapeutic potential. Mol Neurodegener 2023; 18:83. [PMID: 37951933 PMCID: PMC10640762 DOI: 10.1186/s13024-023-00676-7] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/25/2023] [Accepted: 10/30/2023] [Indexed: 11/14/2023] Open
Abstract
Mitochondrial dysfunction is strongly implicated in the etiology of idiopathic and genetic Parkinson's disease (PD). However, strategies aimed at ameliorating mitochondrial dysfunction, including antioxidants, antidiabetic drugs, and iron chelators, have failed in disease-modification clinical trials. In this review, we summarize the cellular determinants of mitochondrial dysfunction, including impairment of electron transport chain complex 1, increased oxidative stress, disturbed mitochondrial quality control mechanisms, and cellular bioenergetic deficiency. In addition, we outline mitochondrial pathways to neurodegeneration in the current context of PD pathogenesis, and review past and current treatment strategies in an attempt to better understand why translational efforts thus far have been unsuccessful.
Collapse
Affiliation(s)
- Martin T Henrich
- Department of Psychiatry and Psychotherapy, Philipps University Marburg, 35039, Marburg, Germany
- Department of Neurology, Philipps University Marburg, 35043, Marburg, Germany
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Wolfgang H Oertel
- Department of Neurology, Philipps University Marburg, 35043, Marburg, Germany
| | - D James Surmeier
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Fanni F Geibl
- Department of Psychiatry and Psychotherapy, Philipps University Marburg, 35039, Marburg, Germany.
- Department of Neurology, Philipps University Marburg, 35043, Marburg, Germany.
- Department of Neuroscience, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
4
|
Naren P, Cholkar A, Kamble S, Khan SS, Srivastava S, Madan J, Mehra N, Tiwari V, Singh SB, Khatri DK. Pathological and Therapeutic Advances in Parkinson's Disease: Mitochondria in the Interplay. J Alzheimers Dis 2023; 94:S399-S428. [PMID: 36093711 PMCID: PMC10473111 DOI: 10.3233/jad-220682] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 08/08/2022] [Indexed: 11/15/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative illness majorly affecting the population between the ages of 55 to 65 years. Progressive dopaminergic neuronal loss and the collective assemblage of misfolded alpha-synuclein in the substantia nigra, remain notable neuro-pathological hallmarks of the disease. Multitudes of mechanistic pathways have been proposed in attempts to unravel the pathogenesis of PD but still, it remains elusive. The convergence of PD pathology is found in organelle dysfunction where mitochondria remain a major contributor. Mitochondrial processes like bioenergetics, mitochondrial dynamics, and mitophagy are under strict regulation by the mitochondrial genome and nuclear genome. These processes aggravate neurodegenerative activities upon alteration through neuroinflammation, oxidative damage, apoptosis, and proteostatic stress. Therefore, the mitochondria have grabbed a central position in the patho-mechanistic exploration of neurodegenerative diseases like PD. The management of PD remains a challenge to physicians to date, due to the variable therapeutic response of patients and the limitation of conventional chemical agents which only offer symptomatic relief with minimal to no disease-modifying effect. This review describes the patho-mechanistic pathways involved in PD not only limited to protein dyshomeostasis and oxidative stress, but explicit attention has been drawn to exploring mechanisms like organelle dysfunction, primarily mitochondria and mitochondrial genome influence, while delineating the newer exploratory targets such as GBA1, GLP, LRRK2, and miRNAs and therapeutic agents targeting them.
Collapse
Affiliation(s)
- Padmashri Naren
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Anjali Cholkar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Suchita Kamble
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Sabiya Samim Khan
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, India
| | - Jitender Madan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, India
| | - Neelesh Mehra
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, India
| | - Vinod Tiwari
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.) Varanasi (U.P.), India
| | - Shashi Bala Singh
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Dharmendra Kumar Khatri
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| |
Collapse
|
5
|
Karvandi MS, Sheikhzadeh Hesari F, Aref AR, Mahdavi M. The neuroprotective effects of targeting key factors of neuronal cell death in neurodegenerative diseases: The role of ER stress, oxidative stress, and neuroinflammation. Front Cell Neurosci 2023; 17:1105247. [PMID: 36950516 PMCID: PMC10025411 DOI: 10.3389/fncel.2023.1105247] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/22/2022] [Accepted: 02/07/2023] [Indexed: 03/08/2023] Open
Abstract
Neuronal loss is one of the striking causes of various central nervous system (CNS) disorders, including major neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and Amyotrophic lateral sclerosis (ALS). Although these diseases have different features and clinical manifestations, they share some common mechanisms of disease pathology. Progressive regional loss of neurons in patients is responsible for motor, memory, and cognitive dysfunctions, leading to disabilities and death. Neuronal cell death in neurodegenerative diseases is linked to various pathways and conditions. Protein misfolding and aggregation, mitochondrial dysfunction, generation of reactive oxygen species (ROS), and activation of the innate immune response are the most critical hallmarks of most common neurodegenerative diseases. Thus, endoplasmic reticulum (ER) stress, oxidative stress, and neuroinflammation are the major pathological factors of neuronal cell death. Even though the exact mechanisms are not fully discovered, the notable role of mentioned factors in neuronal loss is well known. On this basis, researchers have been prompted to investigate the neuroprotective effects of targeting underlying pathways to determine a promising therapeutic approach to disease treatment. This review provides an overview of the role of ER stress, oxidative stress, and neuroinflammation in neuronal cell death, mainly discussing the neuroprotective effects of targeting pathways or molecules involved in these pathological factors.
Collapse
Affiliation(s)
- Mohammad Sobhan Karvandi
- Department of Cell and Molecular Sciences, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | | | - Amir Reza Aref
- Department of Medical Oncology, Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, United States
| | - Majid Mahdavi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
- *Correspondence: Majid Mahdavi
| |
Collapse
|
6
|
Abstract
Parkinson disease (PD) treatment options have conventionally focused on dopamine replacement and provision of symptomatic relief. Current treatments cause undesirable adverse effects, and a large unmet clinical need remains for treatments that offer disease modification and that address symptoms resistant to levodopa. Advances in high-throughput drug screening methods for small molecules, developments in disease modelling and improvements in analytical technologies have collectively contributed to the emergence of novel compounds, repurposed drugs and new technologies. In this Review, we focus on disease-modifying and symptomatic therapies under development for PD. We review cellular therapies and repurposed drugs, such as nilotinib, inosine, isradipine, iron chelators and anti-inflammatories, and discuss how their success in preclinical models has paved the way for clinical trials. We provide an update on immunotherapies and vaccines. In addition, we review non-pharmacological interventions targeting motor symptoms, including gene therapy, adaptive deep brain stimulation (DBS) and optogenetically inspired DBS. Given the many clinical phenotypes of PD, individualization of therapy and precision of treatment are likely to become important in the future.
Collapse
|
7
|
Veryugina NI, Lyashenko EA, Gankina OA. Erectile dysfunction in Parkinson's disease. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 119:51-55. [DOI: 10.17116/jnevro201911909251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/17/2022]
|
8
|
Gao W, He X, Li Y, Wen J. The effects of FK1706 on nerve regeneration and bladder function recovery following an end-to-side neurorrhaphy in rats. Oncotarget 2017; 8:94345-94357. [PMID: 29212232 PMCID: PMC5706878 DOI: 10.18632/oncotarget.21718] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/28/2017] [Accepted: 08/28/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Immunophilin ligands are neuroregenerative agents binding to FK506 binding proteins, by which stimulate recovery of neurons in a variety of injury nerves. FK1706 is a novel immunophilin ligand which has neuroprotective and neuroregenerative effects but without immunosuppressive activity. At present, most reports about FK1706 in ameliorating nerve injury and functional recovery are limited to cavernous nerve injury and erectile function recovery. This study aimed to demonstrate the effects of FK1706 on nerve regeneration and bladder function recovery following an end-to-side neurorrhaphy in rat models. METHOD The numbers of regenerated myelinated axons of the pelvic parasympathetic nerve (PPN) in the three groups' rats (FK1706 + ETS, ETS and control groups) were evaluated. Their intravesical pressure (IVP), S100β and growth associated protein 43 (GAP43) expressions were also compared. RESULTS In FK1706 + ETS group, 90% the rats showed that the frequency of FG labeled neurons was larger than the 3.5 cutoff value, 100% the rats showed that the frequency of FG-FB double-labeled neurons was larger than the 5.5 cutoff value. The average maximum of IVP in FK1706 + ETS group reached 76.3% of the value in control group. Their average number of myelinated axons of regenerated PPN reached 80% of the amount in control group. The nerve regeneration-associated markers data indicated that the expression level of S100β and GAP43 in FK1706 + ETS group was approximately 2-fold higher than that of ETS group (P < 0.05). CONCLUSIONS After end-to-side neurorrhaphy, FK1706 effectively enhanced the nerve regeneration and bladder function recovery.
Collapse
Affiliation(s)
- Wansheng Gao
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Xiangfei He
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Yunlong Li
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| | - Jianguo Wen
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, 450052, China
| |
Collapse
|
9
|
Nambu M, Covel JA, Kapoor M, Li X, Moloney MK, Numa MM, Soltow QA, Trzoss M, Webb P, Webb RR, Mutz M. A calcineurin antifungal strategy with analogs of FK506. Bioorg Med Chem Lett 2017; 27:2465-2471. [PMID: 28412204 DOI: 10.1016/j.bmcl.2017.04.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/14/2017] [Revised: 03/31/2017] [Accepted: 04/01/2017] [Indexed: 10/19/2022]
Abstract
A novel antifungal strategy targeting the inhibition of calcineurin is described. To develop a calcineurin based inhibitor of pathogenic fungi, analogs of FK506 were synthesized that were able to permeate mammalian but not fungal cells. Antagonists in combination with FK506 were not immunosuppressive and retained antifungal activity in A. fumigatus. To reduce the dosage burden of the antagonist, murine oral PK was improved an order of magnitude relative to previous FK506 antagonists.
Collapse
Affiliation(s)
- Mitchell Nambu
- Amplyx Pharmaceuticals, 3210 Merryfield Row, San Diego, CA 92121, United States.
| | - Jonathan A Covel
- Amplyx Pharmaceuticals, 3210 Merryfield Row, San Diego, CA 92121, United States
| | - Mili Kapoor
- Amplyx Pharmaceuticals, 3210 Merryfield Row, San Diego, CA 92121, United States
| | - Xiaoming Li
- Amplyx Pharmaceuticals, 3210 Merryfield Row, San Diego, CA 92121, United States
| | - Molly K Moloney
- Amplyx Pharmaceuticals, 3210 Merryfield Row, San Diego, CA 92121, United States
| | - Mehdi M Numa
- Amplyx Pharmaceuticals, 3210 Merryfield Row, San Diego, CA 92121, United States
| | - Quinlyn A Soltow
- Amplyx Pharmaceuticals, 3210 Merryfield Row, San Diego, CA 92121, United States
| | - Michael Trzoss
- Amplyx Pharmaceuticals, 3210 Merryfield Row, San Diego, CA 92121, United States
| | - Peter Webb
- Amplyx Pharmaceuticals, 3210 Merryfield Row, San Diego, CA 92121, United States
| | - Robert R Webb
- Amplyx Pharmaceuticals, 3210 Merryfield Row, San Diego, CA 92121, United States
| | - Mitchell Mutz
- Amplyx Pharmaceuticals, 3210 Merryfield Row, San Diego, CA 92121, United States.
| |
Collapse
|
10
|
Repair of Neurological Function in Response to FK506 Through CaN/NFATc1 Pathway Following Traumatic Brain Injury in Rats. Neurochem Res 2016; 41:2810-2818. [PMID: 27386875 DOI: 10.1007/s11064-016-1997-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/18/2016] [Revised: 06/25/2016] [Accepted: 06/28/2016] [Indexed: 01/19/2023]
Abstract
Tacrolimus (FK506), an immunophilin ligand, has been widely shown to be neuroprotective in a posttraumatic period. The nuclear factor of activated T cells (NFATc1) pathway plays an important role in regenerating neurological function following traumatic brain injury (TBI), but the precise mechanism underlying FK506-induced repair of neurological functions remains unclear. In the present study, a total of 210 SD rats were enrolled and randomly divided into sham group, TBI group and FK506 group. The rats in the TBI and FK506 groups were inflicted with moderate TBI left lateral fluid percussion impact. A modified neurological severity score (mNSS) system was used to evaluate the severity of effects on nerve function. mNSS levels were significantly lower in the FK506 group than in the TBI group. The zaccumulation of cerebral water content was lower, cerebral Aquaporin 4 (AQP4) mRNA level was lower, the number of growth-associated protein-43 (GAP-43)-positive cells was higher, and the distribution of vesicles containing excitatory neurotransmitters was altered in the injured cortex in the FK506 group. Moreover, the cortical mRNA and serum protein expression levels of interleukin-2 (IL-2) and interferon-γ (IFN-γ) were decreased in FK506 group, especially at 6 h and at 1 day after TBI. At days 1-28 after TBI, the expression of cleaved-caspase 3, which indicates apoptosis, was lower in the FK506 group than in the TBI group. Mechanistically, FK506 significantly down-regulated the mRNA and protein levels of calcium-regulated phosphatase (calcineurin, CaN) and inhibited the activation of NFATc1. These results demonstrate that FK506 relieved inflammatory responses by regulating the NFATc1 signaling pathway and promoting the synaptic reconstruction of neurons and glial cells by regulating cell apoptosis, thereby facilitated improvements in neurological function.
Collapse
|
11
|
Neuroprotective and Therapeutic Strategies against Parkinson's Disease: Recent Perspectives. Int J Mol Sci 2016; 17:ijms17060904. [PMID: 27338353 PMCID: PMC4926438 DOI: 10.3390/ijms17060904] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/25/2016] [Revised: 05/27/2016] [Accepted: 05/30/2016] [Indexed: 12/18/2022] Open
Abstract
Parkinsonism is a progressive motor disease that affects 1.5 million Americans and is the second most common neurodegenerative disease after Alzheimer’s. Typical neuropathological features of Parkinson’s disease (PD) include degeneration of dopaminergic neurons located in the pars compacta of the substantia nigra that project to the striatum (nigro-striatal pathway) and depositions of cytoplasmic fibrillary inclusions (Lewy bodies) which contain ubiquitin and α-synuclein. The cardinal motor signs of PD are tremors, rigidity, slow movement (bradykinesia), poor balance, and difficulty in walking (Parkinsonian gait). In addition to motor symptoms, non-motor symptoms that include autonomic and psychiatric as well as cognitive impairments are pressing issues that need to be addressed. Several different mechanisms play an important role in generation of Lewy bodies; endoplasmic reticulum (ER) stress induced unfolded proteins, neuroinflammation and eventual loss of dopaminergic neurons in the substantia nigra of mid brain in PD. Moreover, these diverse processes that result in PD make modeling of the disease and evaluation of therapeutics against this devastating disease difficult. Here, we will discuss diverse mechanisms that are involved in PD, neuroprotective and therapeutic strategies currently in clinical trial or in preclinical stages, and impart views about strategies that are promising to mitigate PD pathology.
Collapse
|
12
|
Van der Perren A, Macchi F, Toelen J, Carlon MS, Maris M, de Loor H, Kuypers DRJ, Gijsbers R, Van den Haute C, Debyser Z, Baekelandt V. FK506 reduces neuroinflammation and dopaminergic neurodegeneration in an α-synuclein-based rat model for Parkinson's disease. Neurobiol Aging 2015; 36:1559-68. [PMID: 25660193 DOI: 10.1016/j.neurobiolaging.2015.01.014] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 11/25/2014] [Indexed: 10/24/2022]
Abstract
Alpha-synuclein (α-synuclein) is considered a key player in Parkinson's disease (PD), but the exact relationship between α-synuclein aggregation and dopaminergic neurodegeneration remains unresolved. There is increasing evidence that neuroinflammatory processes are closely linked to dopaminergic cell death, but whether the inflammatory process is causally involved in PD or rather reflects secondary consequences of nigrostriatal pathway injury is still under debate. We evaluated the therapeutic effect of the immunophilin ligand FK506 in a rAAV2/7 α-synuclein overexpression rat model. Treatment with FK506 significantly increased the survival of dopaminergic neurons in a dose-dependent manner. No reduction in α-synuclein aggregation was apparent in this time window, but FK506 significantly lowered the infiltration of both T helper and cytotoxic T cells and the number and subtype of microglia and macrophages. These data suggest that the anti-inflammatory properties of FK506 decrease neurodegeneration in this α-synuclein-based PD model, pointing to a causal role of neuroinflammation in the pathogenesis of PD.
Collapse
Affiliation(s)
- Anke Van der Perren
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Flanders, Belgium
| | - Francesca Macchi
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Flanders, Belgium
| | - Jaan Toelen
- Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Flanders, Belgium
| | - Marianne S Carlon
- Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Flanders, Belgium
| | - Michael Maris
- Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Flanders, Belgium
| | - Henriette de Loor
- Division of Nephrology and Renal Transplantation, Department of Microbiology and Immunology, Leuven University Hospital and KU Leuven, Leuven, Belgium
| | - Dirk R J Kuypers
- Division of Nephrology and Renal Transplantation, Department of Microbiology and Immunology, Leuven University Hospital and KU Leuven, Leuven, Belgium
| | - Rik Gijsbers
- Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Flanders, Belgium; Leuven Viral Vector Core, KU Leuven, Leuven, Belgium
| | - Chris Van den Haute
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Flanders, Belgium; Leuven Viral Vector Core, KU Leuven, Leuven, Belgium
| | - Zeger Debyser
- Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Flanders, Belgium; Leuven Viral Vector Core, KU Leuven, Leuven, Belgium
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven, Flanders, Belgium; Leuven Viral Vector Core, KU Leuven, Leuven, Belgium.
| |
Collapse
|
13
|
Abstract
Parkinson disease is an inexorably progressive neurodegenerative disorder. Multiple attempts have been made to establish therapies for Parkinson disease which provide neuroprotection or disease modification-two related, but not identical, concepts. However, to date, none of these attempts have succeeded. Many challenges exist in this field of research, including a complex multisystem disorder that includes dopaminergic and non-dopaminergic features; poorly understood and clearly multifaceted disease pathogenic mechanisms; a lack of reliable animal models; an absence of effective biomarkers of disease state, progression, and target engagement; and the confounding effects of potent symptomatic therapy. In this article, we will review previous, ongoing, and potential future trials designed to alter the progressive course of the disease from the perspective of the targeted underlying pathogenic mechanisms.
Collapse
Affiliation(s)
- Amaal AlDakheel
- />Morton and Gloria Shulman Movement Disorders Clinic and the Edmond J. Safra Program in Parkinson’s Disease, Toronto Western Hospital, University Health Network, Toronto, ON Canada
| | - Lorraine V. Kalia
- />Morton and Gloria Shulman Movement Disorders Clinic and the Edmond J. Safra Program in Parkinson’s Disease, Toronto Western Hospital, University Health Network, Toronto, ON Canada
| | - Anthony E. Lang
- />Movement Disorders Unit, Toronto Western Hospital, 399 Bathurst Street, 7 McLaughlin Wing, Toronto, M5T 2S8 ON Canada
| |
Collapse
|
14
|
Galat A. Functional diversity and pharmacological profiles of the FKBPs and their complexes with small natural ligands. Cell Mol Life Sci 2013; 70:3243-75. [PMID: 23224428 PMCID: PMC11113493 DOI: 10.1007/s00018-012-1206-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/09/2012] [Revised: 10/24/2012] [Accepted: 10/25/2012] [Indexed: 12/25/2022]
Abstract
From 5 to 12 FK506-binding proteins (FKBPs) are encoded in the genomes of disparate marine organisms, which appeared at the dawn of evolutionary events giving rise to primordial multicellular organisms with elaborated internal body plan. Fifteen FKBPs, several FKBP-like proteins and some splicing variants of them are expressed in humans. Human FKBP12 and some of its paralogues bind to different macrocyclic antibiotics such as FK506 or rapamycin and their derivatives. FKBP12/(macrocyclic antibiotic) complexes induce diverse pharmacological activities such as immunosuppression in humans, anticancerous actions and as sustainers of quiescence in certain organisms. Since the FKBPs bind to various assemblies of proteins and other intracellular components, their complexes with the immunosuppressive drugs may differentially perturb miscellaneous cellular functions. Sequence-structure relationships and pharmacological profiles of diverse FKBPs and their involvement in crucial intracellular signalization pathways and modulation of cryptic intercellular communication networks were discussed.
Collapse
Affiliation(s)
- Andrzej Galat
- Commissariat à l'Energie Atomique, Direction des Sciences du Vivant, Institut de Biologie et de Technologies de Saclay, Service d'Ingénierie Moléculaire des Protéines, Bat. 152, 91191, Gif-sur-Yvette Cedex, France.
| |
Collapse
|
15
|
Deleersnijder A, Van Rompuy AS, Desender L, Pottel H, Buée L, Debyser Z, Baekelandt V, Gerard M. Comparative analysis of different peptidyl-prolyl isomerases reveals FK506-binding protein 12 as the most potent enhancer of alpha-synuclein aggregation. J Biol Chem 2011; 286:26687-701. [PMID: 21652707 PMCID: PMC3143632 DOI: 10.1074/jbc.m110.182303] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/21/2010] [Revised: 05/28/2011] [Indexed: 11/06/2022] Open
Abstract
FK506-binding proteins (FKBPs) are members of the immunophilins, enzymes that assist protein folding with their peptidyl-prolyl isomerase (PPIase) activity. Some non-immunosuppressive inhibitors of these enzymes have neuroregenerative and neuroprotective properties with an unknown mechanism of action. We have previously shown that FKBPs accelerate the aggregation of α-synuclein (α-SYN) in vitro and in a neuronal cell culture model for synucleinopathy. In this study we investigated whether acceleration of α-SYN aggregation is specific for the FKBP or even the PPIase family. Therefore, we studied the effect of several physiologically relevant PPIases, namely FKBP12, FKBP38, FKBP52, FKBP65, Pin1, and cyclophilin A, on α-SYN aggregation in vitro and in neuronal cell culture. Among all PPIases tested in vitro, FKBP12 accelerated α-SYN aggregation the most. Furthermore, only FKBP12 accelerated α-SYN fibril formation at subnanomolar concentrations, pointing toward an enzymatic effect. Although stable overexpression of various FKBPs enhanced the aggregation of α-SYN and cell death in cell culture, they were less potent than FKBP12. When FKBP38, FKBP52, and FKBP65 were overexpressed in a stable FKBP12 knockdown cell line, they could not fully restore the number of α-SYN inclusion-positive cells. Both in vitro and cell culture data provide strong evidence that FKBP12 is the most important PPIase modulating α-SYN aggregation and validate the protein as an interesting drug target for Parkinson disease.
Collapse
Affiliation(s)
- Angélique Deleersnijder
- From the Laboratory of Biochemistry and
- Laboratory for Neurobiology and Gene Therapy, K. U. Leuven, Kapucijnenvoer 33, B-3000 Leuven, Flanders, Belgium
| | - Anne-Sophie Van Rompuy
- Laboratory for Neurobiology and Gene Therapy, K. U. Leuven, Kapucijnenvoer 33, B-3000 Leuven, Flanders, Belgium
| | | | - Hans Pottel
- the Laboratory of Biophysics, K. U. Leuven-Kortrijk, Etienne Sabbelaan 53, B-8500 Kortrijk, Flanders, Belgium
| | - Luc Buée
- INSERM, U837, rue Polonovski, F-59000 Lille, France
- Université Lille-Nord de France, UDSL, Faculté de Médecine, Institut de Médecine Prédictive et Recherche Thérapeutique, Université Lille 2, Place de Verdun, F-59045 Lille, France, and
- CHRU, F-59037 Lille Cedex, France
| | - Zeger Debyser
- From the Laboratory of Biochemistry and
- the Laboratory for Molecular Virology and Gene Therapy and
| | - Veerle Baekelandt
- Laboratory for Neurobiology and Gene Therapy, K. U. Leuven, Kapucijnenvoer 33, B-3000 Leuven, Flanders, Belgium
| | - Melanie Gerard
- From the Laboratory of Biochemistry and
- Laboratory for Neurobiology and Gene Therapy, K. U. Leuven, Kapucijnenvoer 33, B-3000 Leuven, Flanders, Belgium
| |
Collapse
|
16
|
Gerard M, Deleersnijder A, Demeulemeester J, Debyser Z, Baekelandt V. Unraveling the role of peptidyl-prolyl isomerases in neurodegeneration. Mol Neurobiol 2011; 44:13-27. [PMID: 21553017 DOI: 10.1007/s12035-011-8184-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/30/2010] [Accepted: 04/14/2011] [Indexed: 02/07/2023]
Abstract
Immunophilins are a family of highly conserved proteins with a peptidyl-prolyl isomerase activity that binds immunosuppressive drugs such as FK506, cyclosporin A, and rapamycin. Immunophilins can be divided into two subfamilies, the cyclophilins, and the FK506 binding proteins (FKBPs). Next to the immunophilins, a third group of peptidyl-prolyl isomerases exist, the parvulins, which do not influence the immune system. The beneficial role of immunophilin ligands in neurodegenerative disease models has been known for more than a decade but remains largely unexplained in terms of molecular mechanisms. In this review, we summarize reported effects of parvulins, immunophilins, and their ligands in the context of neurodegeneration. We focus on the role of FKBP12 in Parkinson's disease and propose it as a novel drug target for therapy of Parkinson's disease.
Collapse
Affiliation(s)
- Melanie Gerard
- Laboratory of Biochemistry, IRC, K.U. Leuven-Kortrijk, Etienne Sabbelaan 53, 8500 Kortrijk, Flanders, Belgium
| | | | | | | | | |
Collapse
|
17
|
|
18
|
Abstract
BACKGROUND Previously we showed that 6-hydroxydopamine lesions of the substantia nigra eliminate corticostriatal LTP and that the neuroimmunolophilin ligand (NIL), GPI-1046, restores LTP. METHODS We used cDNA microarrays to determine what mRNAs may be over- or under-expressed in response to lesioning and/or GPI-1046 treatment. Patch clamp recordings were performed to investigate changes in NMDA channel function before and after treatments. RESULTS We found that 51 gene products were differentially expressed. Among these we found that GPI-1046 treatment up-regulated presenilin-1 (PS-1) mRNA abundance. This finding was confirmed using QPCR. PS-1 protein was also shown to be over-expressed in the striatum of lesioned/GPI-1046-treated rats. As PS-1 has been implicated in controlling NMDA-receptor function and LTP is reduced by lesioning we assayed NMDA mediated synaptic activity in striatal brain slices. The lesion-induced reduction of dopaminergic innervation was accompanied by the near complete loss of NDMA receptor-mediated synaptic transmission between the cortex and striatum. GPI-1046 treatment of the lesioned rats restored NMDA-mediated synaptic transmission but not the dopaminergic innervation. Restoration of NDMA channel function was apparently specific as the sodium channel current density was also reduced due to lesioning but GPI-1046 did not reverse this effect. We also found that restoration of NMDA receptor function was also not associated with either an increase in NMDA receptor mRNA or protein expression. CONCLUSION As it has been previously shown that PS-1 is critical for normal NMDA receptor function, our data suggest that the improvement of excitatory neurotransmission occurs through the GPI-1046-induced up-regulation of PS-1.
Collapse
|
19
|
Inhibition of FK506 binding proteins reduces alpha-synuclein aggregation and Parkinson's disease-like pathology. J Neurosci 2010; 30:2454-63. [PMID: 20164329 DOI: 10.1523/jneurosci.5983-09.2010] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/15/2022] Open
Abstract
alpha-Synuclein (alpha-SYN) is a key player in the pathogenesis of Parkinson's disease (PD). In pathological conditions, the protein is present in a fibrillar, aggregated form inside cytoplasmic inclusions called Lewy bodies. Members of the FK506 binding protein (FKBP) family are peptidyl-prolyl isomerases that were shown recently to accelerate the aggregation of alpha-SYN in vitro. We now established a neuronal cell culture model for synucleinopathy based on oxidative stress-induced alpha-SYN aggregation and apoptosis. Using high-content analysis, we examined the role of FKBPs in aggregation and apoptotic cell death. FK506, a specific inhibitor of this family of proteins, inhibited alpha-SYN aggregation and neuronal cell death in this synucleinopathy model dose dependently. Knockdown of FKBP12 or FKBP52 reduced the number of alpha-SYN aggregates and protected against cell death, whereas overexpression of FKBP12 or FKBP52 accelerated both aggregation of alpha-SYN and cell death. Thus, FK506 likely targets FKBP members in the cell culture model. Furthermore, oral administration of FK506 after viral vector-mediated overexpression of alpha-SYN in adult mouse brain significantly reduced alpha-SYN aggregate formation and neuronal cell death. Our data explain previously described neuroregenerative and neuroprotective effects of immunophilin ligands and validate FKBPs as a novel drug target for the causative treatment of PD.
Collapse
|
20
|
Raju R, Piggott A, Conte M, Tnimov Z, Alexandrov K, Capon R. Nocardiopsins: New FKBP12-Binding Macrolide Polyketides from an Australian Marine-Derived Actinomycete,Nocardiopsissp. Chemistry 2010; 16:3194-200. [DOI: 10.1002/chem.200902933] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/05/2022]
|
21
|
Abstract
The cellular mechanisms underlying neuronal loss and neurodegeneration have been an area of interest in the last decade. Although neurodegenerative diseases such as Alzheimer disease, Parkinson disease, and Huntington disease each have distinct clinical symptoms and pathologies, they all share common mechanisms such as protein aggregation, oxidative injury, inflammation, apoptosis, and mitochondrial injury that contribute to neuronal loss. Although cerebrovascular disease has different causes from the neurodegenerative disorders, many of the same common disease mechanisms come into play following a stroke. Novel therapies that target each of these mechanisms may be effective in decreasing the risk of disease, abating symptoms, or slowing down their progression. Although most of these therapies are experimental, and require further investigation, a few seem to offer promise.
Collapse
Affiliation(s)
- Rawan Tarawneh
- Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO, 63108
- Department of Neurology, Washington University School of Medicine, St Louis, MO, 63108
| | - James E. Galvin
- Alzheimer Disease Research Center, Washington University School of Medicine, St Louis, MO, 63108
- Department of Neurobiology, Washington University School of Medicine, St Louis, MO, 63108
| |
Collapse
|
22
|
Löhle M, Reichmann H. Clinical neuroprotection in Parkinson's disease - still waiting for the breakthrough. J Neurol Sci 2009; 289:104-14. [PMID: 19772974 DOI: 10.1016/j.jns.2009.08.025] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/28/2022]
Abstract
Recent research in the pharmacotherapy of Parkinson's disease (PD) has been able to provide numerous agents for the symptomatic control of motor impairments, but has failed to identify substances capable to slow down or even halt the progression of the disease. In the absence of disease-modifying therapies, affected patients develop marked disability within some years after the onset of motor symptoms, which can be alleviated but eventually not prevented with currently available medical and surgical therapies. Despite promising results from preclinical studies, outcomes of clinical neuroprotection trials have been repeatedly disappointing, which calls for a review of our approach to this topic. This article attempts to explain the need for neuroprotective therapies in PD, discusses results and limitations of previous clinical trials and provides some food for thought for the future research of neuroprotection in PD. Previous experiences from neuroprotection studies may have been discouraging, but also teach us some important lessons for the next generation of preclinical and clinical trials. Firstly, our currently used animal models for PD need to be refined in order to more reliably predict the efficacy of putative neuroprotective agents in subsequent clinical studies. Furthermore, changes in the methodology and design of future neuroprotection trials are required in order to exclude an impact of confounding symptomatic effects on observations. Finally, coordination and concentration of future research on the most promising agents will be necessary in order to accelerate the search for neuroprotective therapies in PD. Just as the pathogenesis of the disease is manifold, it may be this multilateral approach that eventually leads us to a breakthrough in finding neuroprotective agents for PD, if they exist.
Collapse
Affiliation(s)
- Matthias Löhle
- Department of Neurology, Dresden University of Technology, Germany.
| | | |
Collapse
|
23
|
Hayashi H, Campenot RB, Vance DE, Vance JE. Protection of neurons from apoptosis by apolipoprotein E-containing lipoproteins does not require lipoprotein uptake and involves activation of phospholipase Cgamma1 and inhibition of calcineurin. J Biol Chem 2009; 284:29605-13. [PMID: 19717566 DOI: 10.1074/jbc.m109.039560] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/06/2022] Open
Abstract
Apolipoprotein E-containing lipoproteins (LpE) are generated in the central nervous system by glial cells, primarily astrocytes, and are recognized as key players in lipid metabolism and transport in the brain. We previously reported that LpE protect retinal ganglion neurons from apoptosis induced by withdrawal of trophic additives (Hayashi, H., Campenot, R. B., Vance, D. E., and Vance, J. E. (2007) J. Neurosci. 27, 1933-1941). LpE bind to low density lipoprotein receptor-related protein-1 and initiate a signaling pathway that involves activation of protein kinase Cdelta and inhibition of the pro-apoptotic glycogen synthase kinase-3beta. We now show that uptake of LpE is not required for the neuroprotection. Experiments with inhibitors of phospholipase Cgamma1 and RNAi knockdown studies demonstrate that activation of phospholipase Cgamma1 is required for the anti-apoptotic signaling pathway induced by LpE. In addition, the protein phosphatase-2B, calcineurin, is involved in a neuronal death pathway induced by removal of trophic additives, and LpE inhibit calcineurin activation. LpE also attenuate neuronal death caused by oxidative stress. Moreover, physiologically relevant apoE3-containing lipoproteins generated by apoE3 knock-in mouse astrocytes more effectively protect neurons from apoptosis than do apoE4-containing lipoproteins. Because inheritance of the apoE4 allele is the strongest known genetic risk factor for Alzheimer disease, the reduced neuroprotection afforded by apoE4-containing LpE might contribute to the neurodegeneration characteristic of this disease.
Collapse
Affiliation(s)
- Hideki Hayashi
- Priority Organization for Innovation and Excellence, Kumamoto University, Kumamoto 860-8556, Japan
| | | | | | | |
Collapse
|
24
|
Yacoubian TA, Standaert DG. Targets for neuroprotection in Parkinson's disease. BIOCHIMICA ET BIOPHYSICA ACTA 2009; 1792:676-87. [PMID: 18930814 PMCID: PMC2740981 DOI: 10.1016/j.bbadis.2008.09.009] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 08/14/2008] [Revised: 09/16/2008] [Accepted: 09/17/2008] [Indexed: 12/21/2022]
Abstract
Current therapies for Parkinson's disease significantly improve the quality of life for patients suffering from this neurodegenerative disease, yet none of the current therapies has been convincingly shown to slow or prevent the progression of disease. Much has been learned about the pathophysiology of Parkinson's disease in recent years, and these discoveries offer a variety of potential targets for protective therapy. Mechanisms implicated in the disease process include oxidative stress, mitochondrial dysfunction, protein aggregation and misfolding, inflammation, excitotoxicity, and apoptosis. At the same time, the involvement of these diverse processes makes modeling the disease and evaluation of potential treatments difficult. In addition, available clinical tools are limited in their ability to monitor the progression of the disease. In this review, we summarize the different pathogenic mechanisms implicated in Parkinson's disease and neuroprotective strategies targeting these mechanisms currently under clinical study or under preclinical development, with a view towards strategies that seem most promising.
Collapse
Affiliation(s)
- Talene A Yacoubian
- Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | |
Collapse
|
25
|
The development of stroke therapeutics: promising mechanisms and translational challenges. Neuropharmacology 2008; 56:329-41. [PMID: 19007799 DOI: 10.1016/j.neuropharm.2008.10.006] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/08/2008] [Revised: 09/29/2008] [Accepted: 10/06/2008] [Indexed: 12/13/2022]
Abstract
Ischemic stroke is the second most common cause of death worldwide and a major cause of disability. Intravenous thrombolysis with rt-PA remains the only available acute therapy in patients who present within 3h of stroke onset other than the recently approved mechanical MERCI device, substantiating the high unmet need in available stroke therapeutics. The development of successful therapeutic strategies remains challenging, as evidenced by the continued failures of new therapies in clinical trials. However, significant lessons have been learned and this knowledge is currently being incorporated into improved pre-clinical and clinical design. Furthermore, advancements in imaging technologies and continued progress in understanding biological pathways have established a prolonged presence of salvageable penumbral brain tissue and have begun to elucidate the natural repair response initiated by ischemic insult. We review important past and current approaches to drug development with an emphasis on implementing principles of translational research to achieve a rigorous conversion of knowledge from bench to bedside. We highlight current strategies to protect and repair brain tissue with the promise to provide longer therapeutic windows, preservation of multiple tissue compartments and improved clinical success.
Collapse
|
26
|
Current Management of the Cognitive Dysfunction in Parkinson’s Disease: How Far Have We Come? Exp Biol Med (Maywood) 2008; 233:941-51. [DOI: 10.3181/0707-mr-193] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/09/2023] Open
Abstract
Parkinson’s disease (PD) clinical features comprise both motor and nonmotor manifestations. Among the nonmotor complications, dementia is the most important. Approximately 40% of PD patients are affected by cognitive impairment. Remarkably, in addition to age, dementia is an independent predictor of mortality, whereas age at onset of PD and severity of neurological symptoms are not. In this review, I summarize the current knowledge of the pathogenesis of the PD cognitive impairment in relation to the therapies presently accessible and those that could become strategic in the near future. It is hypothesized that patients with PD show two components of cognitive dysfunction (CD): a generalized profile of subcortical dementia (PDsCD), and an overlapped pattern suggesting specific prefrontal damage with CD (PDpFCD). PDsCD is associated with structural neocortical/subcortical changes in the brain (in frontal, parietal, limbic, and temporal lobes, as well as in midbrain structures). In PDpFCD cognitive deficits comprise impairments in neuropsychological tests sensitive for frontal lobe function (discrete elements of episodic and working memory for instance), which are considered to be the consequence of dysfunction in neuronal loops connecting the prefrontal cortex and basal ganglia. Drugs reviewed for targeting PDsCD include: cholinesterase inhibitors, agents with mixed cholinergic and dopaminergic properties, antiglutamatergic drugs, mixed antiglutamatergic/dopaminergic agents; antioxidants and enhancers of mitochondrial functions, and anti-COX-2, as well as other anti-inflammatory mediators. Preliminary studies with vehicles that may target PDpFCD include piribedil, tolcapone, amantadine, and farampator. Additional agents (citicoline and neuroimmuniphilines, among others) will be outlined. A brief overview on neuroprotection and promising new biological advances in PD (deep brain stimulation, stem cells, gene therapy) also will be summarized.
Collapse
|
27
|
Maddess ML, Tackett MN, Ley SV. Total synthesis studies on macrocyclic pipecolic acid natural products: FK506, the antascomicins and rapamycin. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2008; 66:13, 15-186. [PMID: 18416305 DOI: 10.1007/978-3-7643-8595-8_2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 01/04/2023]
Abstract
This chapter derives its inspiration from the challenges presented to total synthesis chemists, by a particular group of macrocyclic pipecolic acid natural products. Although there is considerable emphasis on the completed syntheses of the main characters (FK506 (1), the antascomycins (4 and 5) and rapamycin (7)), the overall complexity of the molecular problem has stimulated a wealth of new knowledge, including the development of novel strategies and the invention of new synthetic methods. The ingenious and innovative approaches to these targets have enabled new generations of analogues, and provided material to further probe the biology of these fascinating molecules. With pharmaceutical application as an immunosuppressant, as well as potential use for the treatment of cancer and neurodegenerative diseases, this family of natural products continues to inspire new and interesting science while providing solutions to healthcare problems of the world.
Collapse
Affiliation(s)
- Matthew L Maddess
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK
| | | | | |
Collapse
|
28
|
Yamaji T, Yamazaki S, Li J, Price RD, Matsuoka N, Mutoh S. FK1706, a novel non-immunosuppressant neurophilin ligand, ameliorates motor dysfunction following spinal cord injury through its neuroregenerative action. Eur J Pharmacol 2008; 591:147-52. [PMID: 18602914 DOI: 10.1016/j.ejphar.2008.06.064] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/26/2008] [Revised: 06/06/2008] [Accepted: 06/12/2008] [Indexed: 11/18/2022]
Abstract
Injured spinal cord axons fail to regenerate in part due to a lack of trophic support. While various methods for replacing neurotrophins have been pursued, clinical uses of these methods face significant barriers. FK1706, a non-immunosuppressant neurophilin ligand, potentiates nerve growth factor signaling, suggesting therapeutic potential for functional deficits following spinal cord injury. Here, we demonstrate that FK1706 significantly improves behavioral outcomes in animal models of spinal cord hemisection and contusion injuries in rats. Furthermore, we show that FK1706 is effective even if administration is delayed until 1 week after injury, suggesting that FK1706 has a reasonable therapeutic time-window. Morphological analysis of injured axons in the dorsal corticospinal tract showed an increase in the radius and perimeter of stained axons, which were reduced by FK1706 treatment, suggesting that axonal swelling and retraction balls observed in injured spinal cord were improved by the neurotrophic effect of FK1706. Taken together, FK1706 improves both behavioral motor function and the underlying morphological changes, suggesting that FK1706 may have therapeutic potential in meeting the significant unmet needs in spinal cord injury.
Collapse
Affiliation(s)
- Takayuki Yamaji
- Pharmacology Research Labs, Astellas Pharma Inc., Tsukuba, Ibaraki, Japan.
| | | | | | | | | | | |
Collapse
|
29
|
Microglial activation is not prevented by tacrolimus but dopamine neuron damage is reduced in a rat model of Parkinson's disease progression. Brain Res 2008; 1216:78-86. [DOI: 10.1016/j.brainres.2008.04.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/29/2008] [Revised: 04/08/2008] [Accepted: 04/10/2008] [Indexed: 11/19/2022]
|
30
|
Rumbaugh JA, Steiner J, Sacktor N, Nath A. Developing neuroprotective strategies for treatment of HIV-associated neurocognitive dysfunction. ACTA ACUST UNITED AC 2008; 2:271-280. [PMID: 19774095 DOI: 10.2217/17469600.2.3.271] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/21/2022]
Abstract
Important advances have been made in recent years in identifying the molecular mechanisms of HIV neuropathogenesis. Defining the pathways leading to HIV dementia has created an opportunity to therapeutically target many steps in the pathogenic process. HIV itself rarely infects neurons, but significant neuronal damage is caused both by viral proteins and by inflammatory mediators produced by the host in response to infection. Highly active antiretroviral therapy (HAART) does not target these mediators of neuronal damage, and the prevalence of HIV-associated neurocognitive dysfunction has actually been rising in the post-HAART era. This review will briefly summarize our current understanding of the mechanisms of HIV-induced neurological disease, and emphasize translation of this basic research into potential clinical applications.
Collapse
Affiliation(s)
- Jeffrey A Rumbaugh
- Johns Hopkins University School of Medicine, Department of Neurology, 600 North Wolfe Street, Baltimore, MD 21287, USA, Tel.: +1 443 287 4656; ;
| | | | | | | |
Collapse
|
31
|
Hung AY, Schwarzschild MA. Clinical trials for neuroprotection in Parkinson??s disease: overcoming angst and futility? Curr Opin Neurol 2007; 20:477-83. [PMID: 17620885 DOI: 10.1097/wco.0b013e32826388d6] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/14/2023]
Abstract
PURPOSE OF REVIEW To summarize recently published results of neuroprotection trials for Parkinson's disease, and discuss them in the context of evolving concepts in clinical study design and animal models. RECENT FINDINGS Despite compelling preclinical evidence from laboratory models suggesting potential neuroprotective benefits, the antioxidant, antiapoptotic, antiexcitotoxic, immunomodulatory and neurotrophic agents studied to date have not shown clear benefit in human studies. The futility study design, an alternative approach focused on efficiently excluding less promising compounds, has been adopted recently to investigate four candidate neuroprotectants. A delayed-start trial design has also been introduced in a study of the monoamine oxidase inhibitor rasagiline, demonstrating a possible neuroprotective effect as well as its clear symptomatic benefit. In parallel with these clinical innovations, preclinical research initiatives are identifying new animal models that more closely resemble the clinical course and pathology of Parkinson's disease. SUMMARY Angst over disappointing results of neuroprotection trials in Parkinson's disease has engendered efforts to refine animal models at one end of the therapeutics pipeline, and to optimize clinical trial design at the other. Building on new insights into the genetics, epidemiology and pathogenesis of Parkinson's disease, these recent improvements in 'translational infrastructure' will enhance the prospects of achieving the critical goal of slowing the progression of disability.
Collapse
Affiliation(s)
- Albert Y Hung
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.
| | | |
Collapse
|
32
|
Newcomb JD, Janowski M, Zigova T, Saporta S, McGrogan MP, Sanberg PR, Willing AE. A comparison of dopaminergic cells from the human NTera2/D1 cell line transplanted into the hemiparkinsonian rat. Life Sci 2007; 81:441-8. [PMID: 17662308 DOI: 10.1016/j.lfs.2007.05.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/24/2006] [Revised: 05/18/2007] [Accepted: 05/29/2007] [Indexed: 11/17/2022]
Abstract
Human NT cells derived from the NTera2/D1 cell line express a dopaminergic phenotype making them an attractive vehicle to supply dopamine to the depleted striatum of the Parkinsonian patient. In vitro, hNT neurons express tyrosine hydroxylase (TH), depending on the length of time they are exposed to retinoic acid. This study compared two populations of hNT neurons that exhibit a high yield of TH+ cells, MI-hNT and DA-hNT. The MI-hNT and DA-hNT neurons were intrastriatally transplanted into the 6-OHDA hemiparkinsonian rat. Amelioration in rotational behavior was measured and immunohistochemistry was performed to identify surviving hNT and TH+ hNT neurons. Results indicated that both MI-hNT and DA-hNT neurons can survive in the striatum, however, neither maintained their dopaminergic phenotype in vivo. Other strategies used in conjunction with hNT cell replacement are likely needed to enhance and maintain the dopamine expression in the grafted cells.
Collapse
Affiliation(s)
- J D Newcomb
- Center for Excellence in Aging and Brain Repair, University of South Florida College of Medicine, 12901 Bruce B. Downs Blvd., Tampa, FL 33612, USA
| | | | | | | | | | | | | |
Collapse
|
33
|
Neuroprotective and antiretroviral effects of the immunophilin ligand GPI 1046. J Neuroimmune Pharmacol 2007; 2:49-57. [PMID: 18040826 DOI: 10.1007/s11481-006-9060-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/14/2006] [Accepted: 12/06/2006] [Indexed: 10/23/2022]
Abstract
HIV infection results in a neurodegenerative disorder for which currently there is no effective therapy available. Currently, available antiretroviral therapy has no impact on the production of early regulatory HIV proteins once the virus is integrated. Of these proteins, Tat was shown to be toxic to neurons. We, thus, used an in vitro neuronal culture system to determine if immunophilin ligands could protect against Tat-induced neurotoxicity. We found that GPI 1046 had potent neuroprotective effects in this model. The compound was able to protect the neurons even though it only partially obliterated Tat-induced oxidative stress in neurons, suggesting that other mechanisms may be important in mediating its neuroprotective effect. Furthermore, GPI 1046 showed inhibition of HIV replication and Tat-mediated long terminal repeat (LTR) activation suggesting that this class of compounds may be worthy of further exploration as a potential treatment for HIV dementia.
Collapse
|
34
|
Valentine H, Chen Y, Guo H, McCormick J, Wu Y, Sezen SF, Hoke A, Burnett AL, Steiner JP. Neuroimmunophilin ligands protect cavernous nerves after crush injury in the rat: new experimental paradigms. Eur Urol 2006; 51:1724-31. [PMID: 17145129 PMCID: PMC2682459 DOI: 10.1016/j.eururo.2006.11.026] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/12/2006] [Accepted: 11/09/2006] [Indexed: 12/13/2022]
Abstract
OBJECTIVES We investigated the effects of the orally bioavailable non-immunosuppressive immunophilin ligand GPI 1046 (GPI) on erectile function and cavernous nerve (CN) histology following unilateral or bilateral crush injury (UCI, BCI, respectively) of the CNs. METHODS Adult male Sprague-Dawley rats were administered GPI 15 mg/kg intraperitoneally (ip) or 30 mg/kg orally (po), FK506 1 mg/kg, ip, or vehicle controls for each route of administration just prior to UCI or BCI and daily up to 7 d following injury. At day 1 or 7 of treatment, erectile function induced by CN electrical stimulation was measured, and electron microscopic analysis of the injured CN was performed. RESULTS Intraperitoneal administration of GPI to rats with injured CN protected erectile function, in a fashion similar to the prototypic immunophilin ligand FK506, compared with vehicle-treated animals (93%+/-9% vs. 70%+/-5% vs. 45%+/-1%, p<0.01, respectively). Oral administration of GPI elicited the same level of significant protection from CN injury. GPI administered po at 30 mg/kg/d, dosing either once daily or four times daily with 7.5 mg/kg, provided nearly complete protection of erectile function. In a more severe BCI model, po administration of GPI maintained erectile function at 24 h after CN injury. Ultrastructural analysis of injured CNs indicated that GPI administered at the time of CN injury prevents degeneration of about 83% of the unmyelinated axons at 7 d after CN injury. CONCLUSIONS The orally administered immunophilin ligand GPI neuroprotects CNs and maintains erectile function in rats under various conditions of CN crush injury.
Collapse
Affiliation(s)
- Heather Valentine
- Department of Research, Guilford Pharmaceuticals Inc (now MGI Pharma), Baltimore, Maryland
| | - Yi Chen
- Department of Research, Guilford Pharmaceuticals Inc (now MGI Pharma), Baltimore, Maryland
| | - Hongzhi Guo
- Department of Research, Guilford Pharmaceuticals Inc (now MGI Pharma), Baltimore, Maryland
| | - Jocelyn McCormick
- Department of Research, Guilford Pharmaceuticals Inc (now MGI Pharma), Baltimore, Maryland
| | - Yong Wu
- Department of Research, Guilford Pharmaceuticals Inc (now MGI Pharma), Baltimore, Maryland
| | - Sena F. Sezen
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ahmet Hoke
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Arthur L. Burnett
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joseph P. Steiner
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Corresponding author. Joseph P. Steiner, PhD, Department of Neurology, The Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD 21287. Tel. (410) 502-3290; Fax: (410) 614-1008. E-mail address: (J. Steiner)
| |
Collapse
|
35
|
Colosimo C, Fabbrini G, Berardelli A. Drug Insight: new drugs in development for Parkinson's disease. ACTA ACUST UNITED AC 2006; 2:600-10. [PMID: 17057747 DOI: 10.1038/ncpneuro0340] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/09/2006] [Accepted: 08/24/2006] [Indexed: 11/08/2022]
Abstract
For many years, levodopa has given most patients with Parkinson's disease excellent symptomatic benefit. This agent does not slow down the progression of the disease, however, and it can induce motor fluctuations and dyskinesias in the long term. The other available antiparkinsonian agents also have drawbacks, and as a consequence research into antiparkinsonian drugs is expected to take new and different directions in the coming years. The most promising approaches include the development of 'neuroprotective' drugs that are capable of blocking or at least slowing down the degenerative process that is responsible for cellular death; 'restorative' strategies intended to restore normal brain function; more-effective agents for replacing dopamine loss; and symptomatic and antidyskinetic drugs that act on neurotransmitters other than dopamine or target brain areas other than the striatum. In this Review, we discuss the numerous drugs in development that target the primary motor disorder in Parkinson's disease.
Collapse
Affiliation(s)
- Carlo Colosimo
- University Department of Neurosciences University of Rome, La Sapienza, Italy.
| | | | | |
Collapse
|
36
|
Abstract
During the past decade, there has been a remarkable progress in our understanding of the biology of Parkinson disease (PD), which has been translated into searching for novel therapy for PD. Much focus is shifted from the development of drugs that only relieve PD symptoms to new generation of remedies that can potentially protect dopaminergic neurons and modify the disease course. Several novel therapeutic approaches have been tested in preclinical experiments and in clinical trials, including molecules targeting on genes involved in the pathogenesis of the disease, neurotrophic factors critical for dopaminergic neuron survival and function, new generation of dopamine receptor agonists that may possess neuroprotective effects, and agents of antioxidation, antiinflammation, and antiapoptosis. The results of these studies will shed new light to our hope that PD can be cured in the future.
Collapse
Affiliation(s)
- Sheng Chen
- Institute of Neurology, Ruijin Hospital, Shanghai 2nd Medical University, Shanghai, China
| | | |
Collapse
|
37
|
Neuromodulatory drugs for the radical prostatectomy patient: Current and future applications. CURRENT SEXUAL HEALTH REPORTS 2006. [DOI: 10.1007/s11930-006-0014-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 10/23/2022]
|
38
|
Hayashi N, Minor TX, Carrion R, Price R, Nunes L, Lue TF. The Effect of FK1706 on Erectile Function Following Bilateral Cavernous Nerve Crush Injury in a Rat Model. J Urol 2006; 176:824-9. [PMID: 16813955 DOI: 10.1016/j.juro.2006.03.071] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/08/2005] [Indexed: 11/23/2022]
Abstract
PURPOSE We investigated the neurotrophic effect of FK1706 on erectile recovery following bilateral cavernous nerve crush injury in a rat model. MATERIALS AND METHODS A total of 28 male Sprague-Dawley rats were randomly divided into 4 equal groups. Seven animals underwent sham operation and subcutaneous vehicle injection, whereas 21 underwent bilateral cavernous nerve crush injury followed by vehicle injection alone, or by low (0.1 mg/kg) or high (1.0 mg/kg) dose FK1706 treatment. Injections were continued 5 days weekly for 8 weeks. Erectile function was then assessed by cavernous nerve electrostimulation and penile tissue was evaluated immunohistochemically. RESULTS No erectile dysfunction was identified in the sham treated group (mean maximal intracavernous pressure +/- SEM 106.8 +/- 6.4 cm H(2)O), whereas nerve injury significantly decreased ICP to 17.9 +/- 7.0 cm H(2)O. FK1706 facilitated neural and erectile recovery in a concentration dependent manner with a mean ICP in the high dose FK treatment group of 80.1 +/- 7.8 cm H(2)O compared with 44.1 +/- 12.9 cm H(2)O in the low dose group. Similar stepwise findings were observed using mean area under the curve data. Sham treated animals showed regular axon sizes and shapes with homogenous GAP-43 and neurofilament staining, whereas injured axons showed irregular shapes, sizes and staining patterns. FK1706 treatment restored axon shape and staining patterns. Injury significantly decreased nicotinamide adenine dinucleotide phosphate staining and FK1706 treatment showed a nonsignificant trend toward increased staining. CONCLUSIONS Bilateral cavernous nerve crush causes reproducible erectile dysfunction, consistent with prior experiments. High dose subcutaneous FK1706 therapy promotes significant neuroregeneration and erectile function recovery.
Collapse
Affiliation(s)
- Narihiko Hayashi
- Knuppe Molecular Urology Laboratory, Department of Urology, University of California-San Francisco, 94143, USA
| | | | | | | | | | | |
Collapse
|
39
|
Edlich F, Weiwad M, Wildemann D, Jarczowski F, Kilka S, Moutty MC, Jahreis G, Lücke C, Schmidt W, Striggow F, Fischer G. The Specific FKBP38 Inhibitor N-(N′,N′-Dimethylcarboxamidomethyl)cycloheximide Has Potent Neuroprotective and Neurotrophic Properties in Brain Ischemia. J Biol Chem 2006; 281:14961-70. [PMID: 16547004 DOI: 10.1074/jbc.m600452200] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/06/2022] Open
Abstract
FK506 and FK506-derived inhibitors of the FK506-binding protein (FKBP)-type peptidylprolyl cis/trans-isomerases (PPIase) display potent neuroprotective and neuroregenerative properties in various neurodegeneration models, showing the importance of neuroimmunophilins as targets for the treatment of acute and chronic neurodegenerative diseases. However, the PPIase activity targeted by active site-directed ligands remains unknown so far. Here we show that neurotrophic FKBP ligands, such as GPI1046 and N-[methyl(ethoxycarbonyl)]cycloheximide, inhibit the calmodulin/Ca(2+) (CaM/Ca(2+))-regulated FKBP38 with up to 80-fold higher affinity than FKBP12. In contrast, the non-neurotrophic rapamycin inhibits FKBP38.CaM/Ca(2+) 500-fold less affine than other neuroimmunophillins. In the context of the high expression of FKBP38 in neuroblastoma cells, these data suggest that FKBP38.CaM/Ca(2+) inhibition can mediate neurotrophic properties of FKBP ligands. The FKBP38-specific cycloheximide derivative, N-(N',N'-dimethylcarboxamidomethyl)cycloheximide (DM-CHX) was synthesized and used in a rat model of transient focal cerebral ischemia. Accordingly, DM-CHX caused neuronal protection as well as neural stem cell proliferation and neuronal differentiation at a dosage of 27.2 mug/kg. These effects were still dominant, if DM-CHX was applied 2-6 h post-insult. In parallel, sustained motor behavior deficits of diseased animals were improved by drug administration, revealing a potential therapeutic relevance. Thus, our results demonstrate that FKBP38 inhibition by DM-CHX regulates neuronal cell death and proliferation, providing a promising strategy for the treatment of acute and/or chronic neurodegenerative diseases.
Collapse
Affiliation(s)
- Frank Edlich
- Max Planck Research Unit for Enzymology of Protein Folding, Weinbergweg 22, D-06120 Halle/Saale, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Hontanilla B, Aubá C, Arcocha J, Gorría O. Nerve Regeneration through Nerve Autografts and Cold Preserved Allografts using Tacrolimus (FK506) in a Facial Paralysis Model: A Topographical and Neurophysiological Study in Monkeys. Neurosurgery 2006; 58:768-79; discussion 768-79. [PMID: 16575341 DOI: 10.1227/01.neu.0000204319.37546.5f] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE Nerve regeneration through cold preserved nerve allografts is demonstrated, and treatment of nerve allografts with FK506 induces better regeneration than other immunosuppressants. We study nerve regeneration through cold preserved nerve allografts temporarily treated with FK506 and compare it with the regeneration obtained using classic nerve autografts in a facial paralysis model in monkeys. METHODS A trunk of the facial nerve on both sides was transected in eight monkeys and immediately repaired with a 3 to 4 cm nerve autograft or allograft. FK506 was administered to the animals of the allograft group for 2 months, and nerve allografts were cold preserved for 3 weeks. At periods of 3, 5, and 8 months after surgery, quantitative electrophysiological assessment and video recordings were performed. At the end of the study, quantitative analysis of neurons in the facial nucleus was carried out, and axons were stereologically counted. RESULTS After the regenerative period, neuronal density was higher in the autograft group. However, distal axonal counts were similar in both groups. Serial electrophysiological recordings and histology of nerve allografts showed that the grafts were partially rejected after cessation of the immunosuppressant. CONCLUSION The regeneration through nerve allografts temporarily treated with FK506 does not achieve the electrophysiological results and neuronal counts achieved with nerve autografts, but axonal collateralization in the allografts induces a similar activation of mimic muscles.
Collapse
Affiliation(s)
- Bernardo Hontanilla
- Department of Plastic and Reconstructive Surgery, Clínica Universitaria, University of Navarra, Pamplona, Spain.
| | | | | | | |
Collapse
|
41
|
Papatsoris AG, Deliveliotis C, Singer C, Papapetropoulos S. Erectile dysfunction in Parkinson's disease. Urology 2006; 67:447-51. [PMID: 16504269 DOI: 10.1016/j.urology.2005.10.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/11/2005] [Revised: 09/14/2005] [Accepted: 10/11/2005] [Indexed: 10/25/2022]
Affiliation(s)
- A G Papatsoris
- 2nd Department of Urology, University of Athens School of Medicine, Athens, Greece
| | | | | | | |
Collapse
|
42
|
Fumagalli F, Racagni G, Riva MA. Shedding light into the role of BDNF in the pharmacotherapy of Parkinson's disease. THE PHARMACOGENOMICS JOURNAL 2006; 6:95-104. [PMID: 16402079 DOI: 10.1038/sj.tpj.6500360] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is a chronic, neurodegenerative disease with a 1% incidence in the population over 55 years of age. Movement impairments represent undoubtedly the hallmark of the disorder; however, extensive evidence implicates cognitive deficits as concomitant peculiar features. Brain-derived neurotrophic factor (BDNF) colocalizes with dopamine neurons in the substantia nigra, where dopaminergic cell bodies are located, and it has recently garnered attention as a molecule crucial for cognition, a function that is also compromised in PD patients. Thus, due to its colocalization with dopaminergic neurons and its role in cognition, BDNF might possess a dual role in PD, both as a neuroprotective molecule, since its inhibition leads to loss of nigral dopaminergic neurons, and as a neuromodulator, as its enhanced expression ameliorates cognitive processes. In this review, we discuss the mechanism of action of established as well as novel drugs for PD with a particular emphasis to those interfering with BDNF biosynthesis.
Collapse
Affiliation(s)
- F Fumagalli
- Department of Pharmacological Sciences, Center of Neuropharmacology, Milan, Italy.
| | | | | |
Collapse
|
43
|
Benn SC, Ay I, Bastia E, Chian RJ, Celia SA, Pepinsky RB, Fishman PS, Brown RH, Francis JW. Tetanus toxin fragment C fusion facilitates protein delivery to CNS neurons from cerebrospinal fluid in mice. J Neurochem 2005; 95:1118-31. [PMID: 16271047 DOI: 10.1111/j.1471-4159.2005.03459.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/29/2022]
Abstract
To improve protein delivery to the CNS following intracerebroventricular administration, we compared the distribution of a human Cu/Zn superoxide dismutase:tetanus toxin fragment C fusion protein (SOD1:TTC) in mouse brain and spinal cord with that of tetanus toxin fragment C (TTC) or human SOD1 (hSOD1) alone, following continuous infusion into the lateral ventricle. Mice infused with TTC or SOD1:TTC showed intense anti-TTC or anti-hSOD1 labeling, respectively, throughout the CNS. In contrast, animals treated with hSOD1 revealed moderate staining in periventricular tissues. In spinal cord sections from animals infused with SOD1:TTC, the fusion protein was found in neuron nuclear antigen-positive (NeuN+) neurons and not glial fibrillary acidic protein-positive (GFAP+) astrocytes. The percentage of NeuN+ ventral horn cells that were co-labeled with hSOD1 antibody was greater in mice treated with SOD1:TTC (cervical cord = 73 +/- 8.5%; lumbar cord = 62 +/- 7.7%) than in mice treated with hSOD1 alone (cervical cord = 15 +/- 3.9%; lumbar cord = 27 +/-4.7%). Enzyme-linked immunosorbent assay for hSOD1 further demonstrated that SOD1:TTC-infused mice had higher levels of immunoreactive hSOD1 in CNS tissue extracts than hSOD1-infused mice. Following 24 h of drug washout, tissue extracts from SOD1:TTC-treated mice still contained substantial amounts of hSOD1, while extracts from hSOD1-treated mice lacked detectable hSOD1. Immunoprecipitation of SOD1:TTC from these extracts using anti-TTC antibody revealed that the recovered fusion protein was structurally intact and enzymatically active. These results indicate that TTC may serve as a useful prototype for development as a non-viral vehicle for improving delivery of therapeutic proteins to the CNS.
Collapse
Affiliation(s)
- Susanna C Benn
- Cecil B. Day Laboratory for Neuromuscular Research, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA
| | | | | | | | | | | | | | | | | |
Collapse
|