1
|
Guo L, Hu H, Jiang N, Yang H, Sun X, Xia H, Ma J, Liu H. Electroacupuncture blocked motor dysfunction and gut barrier damage by modulating intestinal NLRP3 inflammasome in MPTP-induced Parkinson's disease mice. Heliyon 2024; 10:e30819. [PMID: 38774094 PMCID: PMC11107113 DOI: 10.1016/j.heliyon.2024.e30819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 05/01/2024] [Accepted: 05/06/2024] [Indexed: 05/24/2024] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder commonly accompanied by gut dysfunction. EA has shown anti-inflammatory and neuroprotective effects. Here, we aim to explore whether EA can treat Parkinson's disease by restoring the intestinal barrier and modulating NLRP3 inflammasome. We applied 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) to establish a PD mouse model and EA at the GV16, LR3, and ST36 for 12 consecutive days. The open-field test results indicated that EA alleviated depression and behavioral defects, upregulated the expressions of tyrosine hydroxylase (TH) and brain-derived neurotrophic factor (BDNF), and blocked the accumulation of α-synuclein (α-syn) in the midbrain. Moreover, EA blocked the damage to intestinal tissues of PD mice, indicative of suppressed NLRP3 inflammasome activation and increased gut barrier integrity. Notably, the antibiotic-treated mouse experiment validated that the gut microbiota was critical in alleviating PD dyskinesia and intestinal inflammation by EA. In conclusion, this study suggested that EA exhibited a protective effect against MPTP-induced PD by alleviating behavioral defects, reversing the block of motor dysfunction, and improving the gut barrier by modulating intestinal NLRP3 inflammasome. Above all, this study could provide novel insights into the pathogenesis and therapy of PD.
Collapse
Affiliation(s)
- Lei Guo
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, China
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan 430060, China
| | - Haiming Hu
- School of Laboratory Medicine, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, China
| | - Nan Jiang
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430061, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, 430074, China
| | - Huabing Yang
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, China
| | - Xiongjie Sun
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, China
| | - Hui Xia
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, China
| | - Jun Ma
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine, Wuhan 430060, China
| | - Hongtao Liu
- College of Basic Medical Sciences, Hubei University of Chinese Medicine, Huangjiahu West Road 16, Wuhan 430065, China
| |
Collapse
|
2
|
Vanbilloen WJF, Rechberger JS, Anderson JB, Nonnenbroich LF, Zhang L, Daniels DJ. Nanoparticle Strategies to Improve the Delivery of Anticancer Drugs across the Blood-Brain Barrier to Treat Brain Tumors. Pharmaceutics 2023; 15:1804. [PMID: 37513992 PMCID: PMC10383584 DOI: 10.3390/pharmaceutics15071804] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/14/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Primary brain and central nervous system (CNS) tumors are a diverse group of neoplasms that occur within the brain and spinal cord. Although significant advances in our understanding of the intricate biological underpinnings of CNS neoplasm tumorigenesis and progression have been made, the translation of these discoveries into effective therapies has been stymied by the unique challenges presented by these tumors' exquisitely sensitive location and the body's own defense mechanisms (e.g., the brain-CSF barrier and blood-brain barrier), which normally protect the CNS from toxic insult. These barriers effectively prevent the delivery of therapeutics to the site of disease. To overcome these obstacles, new methods for therapeutic delivery are being developed, with one such approach being the utilization of nanoparticles. Here, we will cover the current state of the field with a particular focus on the challenges posed by the BBB, the different nanoparticle classes which are under development for targeted CNS tumor therapeutics delivery, and strategies which have been developed to bypass the BBB and enable effective therapeutics delivery to the site of disease.
Collapse
Affiliation(s)
- Wouter J. F. Vanbilloen
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
- Department of Neurology, Elisabeth-Tweesteden Hospital, 5022 GC Tilburg, The Netherlands
| | - Julian S. Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Jacob B. Anderson
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
- Medical Scientist Training Program, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Leo F. Nonnenbroich
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
- Hopp Children’s Cancer Center Heidelberg (KiTZ), 69120 Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), 69120 Heidelberg, Germany
| | - Liang Zhang
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
| | - David J. Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA (J.S.R.)
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
3
|
Chitu V, Biundo F, Stanley ER. Colony stimulating factors in the nervous system. Semin Immunol 2021; 54:101511. [PMID: 34743926 DOI: 10.1016/j.smim.2021.101511] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 10/23/2021] [Indexed: 01/02/2023]
Abstract
Although traditionally seen as regulators of hematopoiesis, colony-stimulating factors (CSFs) have emerged as important players in the nervous system, both in health and disease. This review summarizes the cellular sources, patterns of expression and physiological roles of the macrophage (CSF-1, IL-34), granulocyte-macrophage (GM-CSF) and granulocyte (G-CSF) colony stimulating factors within the nervous system, with a particular focus on their actions on microglia. CSF-1 and IL-34, via the CSF-1R, are required for the development, proliferation and maintenance of essentially all CNS microglia in a temporal and regional specific manner. In contrast, in steady state, GM-CSF and G-CSF are mainly involved in regulation of microglial function. The alterations in expression of these growth factors and their receptors, that have been reported in several neurological diseases, are described and the outcomes of their therapeutic targeting in mouse models and humans are discussed.
Collapse
Affiliation(s)
- Violeta Chitu
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Fabrizio Biundo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - E Richard Stanley
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
4
|
|
5
|
Xu Y, Tang J, Liu C, Zhao C, Cao S, Yu H, Chen Z, Xie M. MicroPET imaging of vesicular monoamine transporter 2 revealed the potentiation of (+)-dihydrotetrabenazine on MPTP-induced degeneration of dopaminergic neurons. Nucl Med Biol 2021; 96-97:9-18. [PMID: 33647803 DOI: 10.1016/j.nucmedbio.2021.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 01/10/2021] [Accepted: 02/11/2021] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Vesicular monoamine transporter 2 (VMAT2) has been associated with the risk of PD. Genetic reduction of VMAT2 level is reported to increase the vulnerability for dopaminergic neurodegeneration. In this study, by using in vivo microPET imaging with a VMAT2 radioligand [18F]fluoropropyl-(+)-dihydrotetrabenazine ([18F]FP-(+)-DTBZ), we investigated the enhanced role of inhibiting VMAT2 in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced loss of dopaminergic neurons. METHODS The (+)-α-dihydrotetrabenazine ((+)-DTBZ, an inhibitor of VMAT2, 5 mg/kg), or MPTP (low dose (ld): 10 mg/kg, high dose (hd): 30 mg/kg) or both of them were intraperitoneally injected into C57BL/6 mice for 5 or 10 consecutive days. MicroPET imaging with [18F]FP-(+)-DTBZ was performed to test the dopaminergic neuronal integrity. [18F]FP-(+)-DTBZ uptake in striatum was quantified as standardized uptake value (SUV). The pathological changes in the striata and substantia nigra were confirmed by measuring the DA contents and immunohistochemical staining of tyrosine hydroxylase (TH). RESULTS In vivo imaging results showed that the striatal SUVs of both DTBZ&MPTPld and MPTPhd groups were substantially declined compared to the baseline. Moreover, the striatal uptakes of [18F]FP-(+)-DTBZ in DTBZ&MPTPld and MPTPhd groups were obviously lower than the control, DTBZ group and MPTPld group. Notably, the decrease of the striatal uptake in the DTBZ&MPTPld/10d group was more serious than the DTBZ&MPTPld/5d group and comparable to the MPTPhd group. Consistently, the ratios of DA metabolites to DA in DTBZ&MPTPld/10d and MPTPhd mice were significantly increased. The correlation analysis showed that SUVs were highly correlated to the striatal dopaminergic fiber density and TH-positive dopaminergic neuron number in the substantia nigra. CONCLUSIONS MicroPET brain imaging with [18F]FP-(+)-DTBZ noninvasively revealed that (+)-DTBZ co-administration significantly aggravated the neurotoxicity of MPTP to dopaminergic neurons, suggesting that inhibition of VMAT2 may be related to the pathogenesis of PD and tracing VMAT2 activity with PET imaging is of potential value in monitoring PD progression.
Collapse
Affiliation(s)
- Yingjiao Xu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jie Tang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
| | - Chunyi Liu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
| | - Chao Zhao
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
| | - Shanshan Cao
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
| | - Huixin Yu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China
| | - Zhengping Chen
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China.
| | - Minhao Xie
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu 214063, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
6
|
Zhang G, Jiang B, Wu C, Liu Y, He Y, Huang X, Chen W, Xi K, Guo H, Zhao X, Jia X. Thin platelet-like COF nanocomposites for blood brain barrier transport and inhibition of brain metastasis from renal cancer. J Mater Chem B 2021; 8:4475-4488. [PMID: 32365151 DOI: 10.1039/d0tb00724b] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Effective treatment of brain metastases is hindered by the blood-brain barrier (BBB) and the rapid development of resistance to drug therapy. Moreover, the clinical application of general formulations is hampered by biological barriers and biological elimination. To tackle this challenge, we report a feasible approach for the assembly of polymer-covalent organic framework (COF) nanocomposites into 150 nm thin platelets as a drug delivery vehicle for enhanced retention in brain tumours. Using intravital imaging, we demonstrate that these polymer-COF nanocomposites are able to traverse the BBB in mice and achieve direct tumour accumulation in intracranial orthotopic models of brain metastasis from renal cancer (BMRC). These nanocomposites can target brain tumour cells and respond to tumour microenvironmental characteristics, including acidic and redox conditions. Intracranial tumour acidity triggers the breakdown of the nanoassemblies to polymer-COF nanocomposites due to the presence of borate bonds. Furthermore, in vivo studies on the nanocomposites showed enhanced brain tumour-targeting efficiency and therapeutic effects compared to those of free-drug dosing. Mice treated with drug-loaded polymer-COF nanocomposites also show protection from systemic drug toxicity and improved survival, demonstrating the preclinical potential of this nanoscale platform to deliver novel combination therapies to BMRC and other central nervous system (CNS) tumours.
Collapse
Affiliation(s)
- Guiyang Zhang
- School of Chemistry and Chemical Engineering, Nanjing University, Jiangsu 210023, China. and Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Bo Jiang
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, P. R. China.
| | - Chunyong Wu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China
| | - Yanfeng Liu
- School of Chemistry and Chemical Engineering, Nanjing University, Jiangsu 210023, China.
| | - Yidan He
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China
| | - Xin Huang
- School of Chemistry and Chemical Engineering, Nanjing University, Jiangsu 210023, China.
| | - Wei Chen
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, P. R. China.
| | - Kai Xi
- School of Chemistry and Chemical Engineering, Nanjing University, Jiangsu 210023, China.
| | - Hongqian Guo
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, P. R. China.
| | - Xiaozhi Zhao
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, 321 Zhongshan Road, Nanjing, 210008, P. R. China.
| | - Xudong Jia
- School of Chemistry and Chemical Engineering, Nanjing University, Jiangsu 210023, China.
| |
Collapse
|
7
|
Fan S, Yin Q, Li D, Ma J, Li L, Chai S, Guo H, Yang Z. Anti-neuroinflammatory effects of Eucommia ulmoides Oliv. In a Parkinson's mouse model through the regulation of p38/JNK-Fosl2 gene expression. JOURNAL OF ETHNOPHARMACOLOGY 2020; 260:113016. [PMID: 32464317 DOI: 10.1016/j.jep.2020.113016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 05/15/2020] [Accepted: 05/24/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Eucommia ulmoides Oliv., a Chinese medicinal herb called "Duzhong" from the bark of Eucommia ulmoides Oliv., has been shown to possess significant protective effects in Parkinson's disease (PD). However, the molecular mechanism remains unclear. AIM OF THE STUDY In this study, we explored the anti-neuroinflammatory mechanisms of Duzhong on the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model to elucidate the traditional medical theories with modern pharmacological methods and to provide a reference for further clarifying its mechanisms of action. MATERIALS AND METHODS The representative components in Duzhong extract were identified by UPLC-Q-TOF/MS. Male C57BL/6J mice were intraperitoneally injected with MPTP to establish an in vivo PD model. The pole, rotarod, and grip strength tests were performed to evaluate the motor coordination ability of the PD mice. HPLC-ECD was used to detect the striatal levels of dopamine (DA), 3,4- dihydroxyphenylacetic acid (DOPAC) and homovanillic acid (HVA). The expression of tyrosine hydroxylase (TH) was studied by immunohistochemistry (IHC) and Western blot assays. ELISA and Q-PCR were used examined the levels of proinflammatory cytokines in the serum and midbrain, respectively. Whole-transcriptome analysis of the midbrain was performed to explore the therapeutic effect of Duzhong on PD mice, and Q-PCR was then used to validate the differential gene expression changes in the PD mice treated with Duzhong. RESULTS Ten compounds were identified from Duzhong extract. Duzhong significantly alleviated the behavioral impairments and dopaminergic neuron degeneration of PD mice, and inhibited the expression of proinflammatory cytokines. Whole-transcriptome analysis revealed nine oppositely regulated genes, and the Fosl2 gene was consistent with the trend of observed by RNA-seq. Furthermore, Duzhong downregulated mRNA expression of p38 and JNK, which are key upstream genes of Fosl2. CONCLUSIONS Duzhong has promising therapeutic potential in PD mice, and its molecular mechanism is mediated by downregulating p38/JNK-Fosl2 gene expression to alleviate neuroinflammation.
Collapse
Affiliation(s)
- Shanshan Fan
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, #10 Boyanghu Road, Jinghai District, Tianjin, 301617, China
| | - Qingsheng Yin
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, #10 Boyanghu Road, Jinghai District, Tianjin, 301617, China
| | - Dongna Li
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, #10 Boyanghu Road, Jinghai District, Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jing Ma
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, #10 Boyanghu Road, Jinghai District, Tianjin, 301617, China
| | - Lili Li
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, #10 Boyanghu Road, Jinghai District, Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shiwei Chai
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Hong Guo
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, #10 Boyanghu Road, Jinghai District, Tianjin, 301617, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Zhen Yang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, #10 Boyanghu Road, Jinghai District, Tianjin, 301617, China.
| |
Collapse
|
8
|
Ghahari L, Safari M, Rahimi Jaberi K, Jafari B, Safari K, Madadian M. Mesenchymal Stem Cells with Granulocyte Colony-Stimulating Factor Reduce Stress Oxidative Factors in Parkinson's Disease. IRANIAN BIOMEDICAL JOURNAL 2019. [PMID: 31677610 PMCID: PMC6984711 DOI: 10.29252/ibj.24.2.89] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background: Recent studies have shown that BMSCs have a putative ability to promote neurogenesis and produce behavioral and functional improvement. Our previous study demonstrated that co-treatment of G-CSF and BMSCs have beneficial effects on Parkinson's models. The main purpose of this research was to investigate the effects of these two factors on oxidative stress factors in the brain of Parkinson's rat. Methods: Adult male Wistar rats (weighing 200–250 g) were used and randomly divided into five groups of seven each. To create the Parkinson's model, 6-OHDA was injected into the left SNpc. The BMSCs (2 × 106) and G-CSF (75 µg/kg) were used for treatment after creating the PD model. After four weeks, the brains of rats were removed and processed for immunohistochemical studies, such as TH-positive neurons as well as analysis of oxidative stress factors. Results: The results showed that the injected BMSCs could cross the BBB. The injected cells are also able to settle in different areas of the brain. Analyses of the brain oxidative stress factors showed that G-CSF and BMSCs reduced the expression of MDA and induced the activity of SOD, GSH-Px, and FRAP. Conclusion: Co-administration of G-CSF and BMSCs reduced the expression of pro-inflammatory cytokines and induced the activity of antioxidant enzymes; however, neurogenesis increased in the brain.
Collapse
Affiliation(s)
- Laya Ghahari
- Department of Anatomy, AJA University of Medical Sciences, Tehran, Iran
| | - Manouchehr Safari
- Nervous System Stem Cells Research Center, Semnan university of Medical Sciences, Semnan, Iran
| | - Khojaste Rahimi Jaberi
- Nervous System Stem Cells Research Center, Semnan university of Medical Sciences, Semnan, Iran
| | | | - Katayoun Safari
- Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mahmoodreza Madadian
- School of Pharmacology, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
9
|
Su J, Wang H, Yang Y, Wang J, Li H, Huang D, Huang L, Bai X, Yu M, Fei J, Huang F. RESP18 deficiency has protective effects in dopaminergic neurons in an MPTP mouse model of Parkinson's disease. Neurochem Int 2018; 118:195-204. [DOI: 10.1016/j.neuint.2018.06.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 06/24/2018] [Accepted: 06/27/2018] [Indexed: 10/28/2022]
|
10
|
El-Esawy R, Balaha M, Kandeel S, Hadya S, El-Rahman MNA. Filgrastim (G-CSF) ameliorates Parkinsonism l -dopa therapy’s drawbacks in mice. BASAL GANGLIA 2018; 13:17-26. [DOI: 10.1016/j.baga.2018.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
11
|
Azmy MS, Menze ET, El-Naga RN, Tadros MG. Neuroprotective Effects of Filgrastim in Rotenone-Induced Parkinson's Disease in Rats: Insights into its Anti-Inflammatory, Neurotrophic, and Antiapoptotic Effects. Mol Neurobiol 2018; 55:6572-6588. [PMID: 29327204 DOI: 10.1007/s12035-017-0855-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 12/20/2017] [Indexed: 12/13/2022]
Abstract
All current treatments of Parkinson's disease (PD) focus on enhancing the dopaminergic effects and providing symptomatic relief; however, they cannot delay the disease progression. Filgrastim, a recombinant methionyl granulocyte colony-stimulating factor, demonstrated neuroprotection in many neurodegenerative and neurological diseases. This study aimed to assess the neuroprotective effects of filgrastim in rotenone-induced rat model of PD and investigate the potential underlying mechanisms of filgrastim actions. The effects of two doses of filgrastim (20 and 40 μg/kg) on spontaneous locomotion, catalepsy, body weight, histology, and striatal dopamine (DA) content, as well as tyrosine hydroxylase (TH) and α-synuclein expression, were evaluated. Then, the effective dose was further tested for its potential anti-inflammatory, neurotrophic, and antiapoptotic effects. Filgrastim (40 μg/kg) prevented rotenone-induced motor deficits, weight reduction, striatal DA depletion, and histological damage. Besides, it significantly inhibited rotenone-induced decrease in TH expression and increase in α-synuclein immunoreactivity in the midbrains and striata of the rats. These effects were associated with reduction of rotenone-induced neuroinflammation, apoptosis, and brain-derived neurotrophic factor depletion. Collectively, these results suggest that filgrastim might be a good candidate for management of PD.
Collapse
Affiliation(s)
- Mariama S Azmy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Esther T Menze
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Reem N El-Naga
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Mariane G Tadros
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
12
|
Banks WA, Kovac A, Morofuji Y. Neurovascular unit crosstalk: Pericytes and astrocytes modify cytokine secretion patterns of brain endothelial cells. J Cereb Blood Flow Metab 2018; 38:1104-1118. [PMID: 29106322 PMCID: PMC5998993 DOI: 10.1177/0271678x17740793] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Crosstalk among brain endothelial cells (BECs), pericytes, and astrocytes occurs by way of soluble factors, including cytokines. Here, we studied cytokine secretion from both mouse BEC monocultures and tri-cultured with pericytes and astrocytes. Four cytokines were constitutively secreted by BEC monolayers, 12 by LPS-stimulated BECs, 10 by tri-cultures, and 14 by LPS-stimulated tri-cultures. Cytokine levels were generally higher with either LPS stimulation or tri-culture when compared to monocultures and highest in tri-cultures stimulated by LPS. LPS-stimulated secretions fell into eight patterns as categorized by the polarization of cytokine secretions. To determine the cellular origin of cytokine increases in tri-cultures, we cultured mouse BECs with human pericytes and astrocytes and measured cytokines in species-specific assays. Thus, cytokines detected in the human immunoassay were from pericytes/astrocytes and those detected in the mouse immunoassay were from BECs. Several unique patterns were thus found. For example, TNF-alpha was only of pericyte/astrocyte origin; granulocyte colony-stimulating factor was only of BEC origin; IL-6, MCP-1, and GM-CSF of astrocyte/pericyte origin were found in both the luminal and abluminal chambers, suggesting the presence of brain-to-blood transporters. We conclude that crosstalk influences cytokine secretion under constitutive and stimulated conditions from both BECs and pericytes/astrocytes.
Collapse
Affiliation(s)
- William A Banks
- 1 Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA.,2 Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Andrej Kovac
- 3 Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Yoichi Morofuji
- 4 Department of Neurosurgery, University of Nagasaki, Nagasaki, Japan
| |
Collapse
|
13
|
Dong J, Cui Y, Li S, Le W. Current Pharmaceutical Treatments and Alternative Therapies of Parkinson's Disease. Curr Neuropharmacol 2016; 14:339-55. [PMID: 26585523 PMCID: PMC4876590 DOI: 10.2174/1570159x14666151120123025] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 07/16/2015] [Accepted: 10/09/2015] [Indexed: 01/01/2023] Open
Abstract
Over the decades, pharmaceutical treatments, particularly dopaminergic (DAergic) drugs have been considered as the main therapy against motor symptoms of Parkinson's disease (PD). It is proposed that DAergic drugs in combination with other medications, such as monoamine oxidase type B inhibitors, catechol-O-methyl transferase inhibitors, anticholinergics and other newly developed non-DAergic drugs can make a better control of motor symptoms or alleviate levodopa-induced motor complications. Moreover, non-motor symptoms of PD, such as cognitive, neuropsychiatric, sleep, autonomic and sensory disturbances caused by intrinsic PD pathology or drug-induced side effects, are gaining increasing attention and urgently need to be taken care of due to their impact on quality of life. Currently, neuroprotective therapies have been investigated extensively in pre-clinical studies, and some of them have been subjected to clinical trials. Furthermore, non-pharmaceutical treatments, including deep brain stimulation (DBS), gene therapy, cell replacement therapy and some complementary managements, such as Tai chi, Yoga, traditional herbs and molecular targeted therapies have also been considered as effective alternative therapies to classical pharmaceutics. This review will provide us updated information regarding the current drugs and non-drugs therapies for PD.
Collapse
Affiliation(s)
| | | | | | - Weidong Le
- Neurology and Director of Center for Translational Research of Neurological Diseases, 1st Affiliated Hospital, Dalian Medical University, Dalian 116021, Liaoning Province, China.
| |
Collapse
|
14
|
Tsai ST, Chu SC, Liu SH, Pang CY, Hou TW, Lin SZ, Chen SY. Neuroprotection of Granulocyte Colony-Stimulating Factor for Early Stage Parkinson's Disease. Cell Transplant 2016; 26:409-416. [PMID: 27938485 DOI: 10.3727/096368916x694247] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Parkinson's disease (PD) is a slowly progressive neurodegenerative disease. Both medical and surgical choices provide symptomatic treatment. Granulocyte colony-stimulating factor (G-CSF), a conventional treatment for hematological diseases, has demonstrated its effectiveness in acute and chronic neurological diseases through its anti-inflammatory and antiapoptosis mechanisms. Based on previous in vitro and in vivo studies, we administered a lower dose (3.3 μg/kg) G-CSF injection for 5 days and six courses for 1 year in early-stage PD patients as a phase I trial. The four PD patient's mean unified PD rating scale motor scores in medication off status remained stable from 23 before the first G-CSF injection to 22 during the 2-year follow-up. 3,4-Dihydroxy-6-18F-fluoro-l-phenylalanine (18F-DOPA) positron emission tomography (PET) studies also revealed an annual 3.5% decrease in radiotracer uptake over the caudate nucleus and 7% in the putamen, both slower than those of previous reports of PD. Adverse effects included transient muscular-skeletal pain, nausea, vomiting, and elevated liver enzymes. Based on this preliminary report, G-CSF seems to alleviate disease deterioration for early stage PD patients. The effectiveness of G-CSF was possibly due to its amelioration of progressive dopaminergic neuron degeneration.
Collapse
|
15
|
Gasco S, Rando A, Zaragoza P, García-Redondo A, Calvo AC, Osta R. Hematopoietic stem and progenitor cells as novel prognostic biomarkers of longevity in a murine model for amyotrophic lateral sclerosis. Am J Physiol Cell Physiol 2016; 311:C910-C919. [PMID: 27681176 DOI: 10.1152/ajpcell.00081.2016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Accepted: 09/21/2016] [Indexed: 11/22/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with a difficult diagnosis and prognosis. In this regard, new and more reliable biomarkers for the disease are needed. We propose peripheral blood, and, more specifically, the hematopoietic stem and progenitor cells (HSPCs) as potential prognostic biomarkers in the SOD1G93A murine model of ALS. We accurately and serially studied three HSPCs-hematopoietic stem cells (HSCs), common lymphoid progenitors (CLPs), and common myeloid progenitors (CMPs)-in both control and SOD1G93A mice along the disease's progression by RT-PCR and flow cytometry analysis. We found interesting differences for every HSPC type in the transgenic mice compared with the control mice at every time point selected, as well as differences along the disease course. The results showed a maintained compensatory increase of HSCs along disease progression. However, the downregulated levels of CLPs and CMPs suggested an exit of these cell populations to the peripheral tissues, probably due to their supporting role to the damaged tissues. In addition, a positive correlation of the percentage of CLPs and CMPs with the longevity was found, as well as a positive correlation of HSCs and CMPs with motor function and weight, thus reinforcing the idea that HSPCs play a relevant role in the longevity of the SOD1G93A mice. On the basis of these results, both CLPs and CMPs could be considered prognostic biomarkers of longevity in this animal model, opening the door to future studies in human patients for their potential clinical use.
Collapse
Affiliation(s)
- Samanta Gasco
- Laboratorio de Genética Bioquímica, Veterinary Faculty of Zaragoza, Instituto Agroalimentario de Aragón, Health Research Institute of Aragon, University of Zaragoza, Zaragoza, Spain; and
| | - Amaya Rando
- Laboratorio de Genética Bioquímica, Veterinary Faculty of Zaragoza, Instituto Agroalimentario de Aragón, Health Research Institute of Aragon, University of Zaragoza, Zaragoza, Spain; and
| | - Pilar Zaragoza
- Laboratorio de Genética Bioquímica, Veterinary Faculty of Zaragoza, Instituto Agroalimentario de Aragón, Health Research Institute of Aragon, University of Zaragoza, Zaragoza, Spain; and
| | - Alberto García-Redondo
- Biochemistry Department, Centre for Biomedical Network Research on Rare Diseases, Health Research Institute, October 12th Hospital, Madrid, Spain
| | - Ana Cristina Calvo
- Laboratorio de Genética Bioquímica, Veterinary Faculty of Zaragoza, Instituto Agroalimentario de Aragón, Health Research Institute of Aragon, University of Zaragoza, Zaragoza, Spain; and
| | - Rosario Osta
- Laboratorio de Genética Bioquímica, Veterinary Faculty of Zaragoza, Instituto Agroalimentario de Aragón, Health Research Institute of Aragon, University of Zaragoza, Zaragoza, Spain; and
| |
Collapse
|
16
|
Komnig D, Imgrund S, Reich A, Gründer S, Falkenburger BH. ASIC1a Deficient Mice Show Unaltered Neurodegeneration in the Subacute MPTP Model of Parkinson Disease. PLoS One 2016; 11:e0165235. [PMID: 27820820 PMCID: PMC5098794 DOI: 10.1371/journal.pone.0165235] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 09/15/2016] [Indexed: 12/21/2022] Open
Abstract
Inflammation contributes to the death of dopaminergic neurons in Parkinson disease and can be accompanied by acidification of extracellular pH, which may activate acid-sensing ion channels (ASIC). Accordingly, amiloride, a non-selective inhibitor of ASIC, was protective in an acute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson disease. To complement these findings we determined MPTP toxicity in mice deficient for ASIC1a, the most common ASIC isoform in neurons. MPTP was applied i.p. in doses of 30 mg per kg on five consecutive days. We determined the number of dopaminergic neurons in the substantia nigra, assayed by stereological counting 14 days after the last MPTP injection, the number of Nissl positive neurons in the substantia nigra, and the concentration of catecholamines in the striatum. There was no difference between ASIC1a-deficient mice and wildtype controls. We are therefore not able to confirm that ASIC1a are involved in MPTP toxicity. The difference might relate to the subacute MPTP model we used, which more closely resembles the pathogenesis of Parkinson disease, or to further targets of amiloride.
Collapse
Affiliation(s)
- Daniel Komnig
- Department of Neurology, RWTH Aachen University, Aachen, Germany
| | - Silke Imgrund
- Institute of Physiology, RWTH Aachen University, Aachen, Germany
| | - Arno Reich
- Department of Neurology, RWTH Aachen University, Aachen, Germany
| | - Stefan Gründer
- Institute of Physiology, RWTH Aachen University, Aachen, Germany
| | - Björn H. Falkenburger
- Department of Neurology, RWTH Aachen University, Aachen, Germany
- JARA BRAIN Institute II, Jülich and Aachen, Germany
- * E-mail:
| |
Collapse
|
17
|
Komnig D, Schulz JB, Reich A, Falkenburger BH. Mice lacking Faim2 show increased cell death in the MPTP mouse model of Parkinson disease. J Neurochem 2016; 139:848-857. [PMID: 27638043 DOI: 10.1111/jnc.13847] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 09/12/2016] [Accepted: 09/13/2016] [Indexed: 12/24/2022]
Abstract
The death receptor Fas/CD95 mediates apoptotic cell death in response to external stimuli. In neurons, Fas-induced apoptosis is prevented by Fas-apoptotic inhibitory molecule 2 (Faim2). Mice lacking Faim2 showed increased neurodegeneration in animal models of stroke and bacterial meningitis. We therefore tested the relevance of Faim2 in a classical animal model of Parkinson disease and determined the toxicity of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) in Faim2-deficient mice. Without MPTP treatment, there was no difference in the dopaminergic system between Faim2-deficient mice and control mice. MPTP was applied i.p. in doses of 30 mg per kg on five consecutive days. Fourteen days after the last MPTP injection, the number of dopaminergic neurons in the lateral substantia nigra, assayed by stereological counting, was reduced by 39% in control mice and 53% in Faim2-deficient mice. The density of dopaminergic fibers in the dorsal striatum was reduced by 36% in control mice and 69% in Faim2-deficient mice, in the ventral striatum 44% in control mice and 76% in Faim2-deficient mice. Fiber density recovered at 90 days after MPTP with similar density in both groups. Striatal catecholamine levels were reduced by 81-84% in both groups and recovered at 90 days. Faim2 expression was documented in mouse midbrain using quantitative reverse transcription-PCR (qRT-PCR) and found decreased after MPTP administration. Taken together, our findings demonstrate increased degeneration of dopaminergic neurons with Faim2 deficiency, indicating that Fas-induced apoptosis contributes to cell death in the MPTP mouse model. Along with the decreased expression of Faim2 after MPTP, this finding indicates that boosting Faim2 function might represent a therapeutic strategy for Parkinson disease.
Collapse
Affiliation(s)
- Daniel Komnig
- Department of Neurology, RWTH University Aachen, Aachen, Germany
| | - Jörg B Schulz
- Department of Neurology, RWTH University Aachen, Aachen, Germany.,JARA-Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, Aachen, Germany
| | - Arno Reich
- Department of Neurology, RWTH University Aachen, Aachen, Germany
| | - Björn H Falkenburger
- Department of Neurology, RWTH University Aachen, Aachen, Germany.,JARA-Institute Molecular Neuroscience and Neuroimaging, Forschungszentrum Jülich GmbH and RWTH Aachen University, Aachen, Germany
| |
Collapse
|
18
|
Li H, Linjuan-Li, Wang Y. G-CSF improves CUMS-induced depressive behaviors through downregulating Ras/ERK/MAPK signaling pathway. Biochem Biophys Res Commun 2016; 479:827-832. [PMID: 27680311 DOI: 10.1016/j.bbrc.2016.09.123] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 09/24/2016] [Indexed: 12/13/2022]
Abstract
Neuronal plasticity in hippocampal neurons is closely related to memory, mood and behavior as well as in the development of depression. Granulocyte colony-stimulating factor (G-CSF) can promote neuronal plasticity and enhance motor skills. However, the function of G-CSF in depression remains poorly understood. In this study, we explored the biological role and potential molecular mechanism of G-CSF on depression-like behaviors. Our results showed that G-CSF was significantly downregulated in the hippocampus of chronic unexpected mild stress (CUMS) rats. Administration of G-CSF significantly reversed CUMS-induced depression-like behaviors in the open field test (OFT), sucrose preference test (SPT) and forced swimming test (FST). Moreover, G-CSF upregulated the expression of synaptic-associated proteins including polysialylated form of neural cell adhesion molecule (PSA-NCAM), synaptophysin (SYN), and postsynaptic density protein 95 (PSD-95) in the hippocampus and G-CSF significantly increased cell viability rate of hippocampal neurons in vitro. Further studies indicated that the renin-angiotensin system (Ras)/extracellular signal-regulated kinase (ERK)/p38 mitogen-activated protein kinase (MAPK) signaling pathways was involved in the regulation of G-CSF on depressive-like behaviors and neuronal plasticity in CUMS rats. Taken together, our results showed that G-CSF improves depression-like behaviors via inhibiting Ras/ERK/MAPK signaling pathways. Our study suggests that G-CSF may be a promising therapeutic strategy for the treatment of depression.
Collapse
Affiliation(s)
- Hui Li
- Department of Psychiatry and Psychology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Linjuan-Li
- Department of General Internal Medicine, Affiliated Hospital of Yanan University, Yanan, 716000, China; Department of Physiology and Pathophysiology, Health Science Center of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Yaping Wang
- Department of Psychiatry and Psychology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.
| |
Collapse
|
19
|
Kumar AS, Jagadeeshan S, Subramanian A, Chidambaram SB, Surabhi RP, Singhal M, Bhoopalan H, Sekar S, Pitani RS, Duvuru P, Venkatraman G, Rayala SK. Molecular Mechanism of Regulation of MTA1 Expression by Granulocyte Colony-stimulating Factor. J Biol Chem 2016; 291:12310-21. [PMID: 27044752 PMCID: PMC4933278 DOI: 10.1074/jbc.m115.707224] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 03/21/2016] [Indexed: 11/06/2022] Open
Abstract
Parkinson disease (PD) is a neurodegenerative disorder with loss of dopaminergic neurons of the brain, which results in insufficient synthesis and action of dopamine. Metastasis-associated protein 1 (MTA1) is an upstream modulator of tyrosine hydroxylase (TH), the rate-limiting enzyme in dopamine synthesis, and hence MTA1 plays a significant role in PD pathogenesis. To impart functional and clinical significance to MTA1, we analyzed MTA1 and TH levels in the substantia nigra region of a large cohort of human brain tissue samples by Western blotting, quantitative PCR, and immunohistochemistry. Our results showed that MTA1 and TH levels were significantly down-regulated in PD samples as compared with normal brain tissue. Correspondingly, immunohistochemistry analysis for MTA1 in substantia nigra sections revealed that 74.1% of the samples had a staining intensity of <6 in the PD samples as compared with controls, 25.9%, with an odds ratio of 8.54. Because of the clinical importance of MTA1 established in PD, we looked at agents to modulate MTA1 expression in neuronal cells, and granulocyte colony-stimulating factor (G-CSF) was chosen, due to its clinically proven neurogenic effects. Treatment of the human neuronal cell line KELLY and acute 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model with G-CSF showed significant induction of MTA1 and TH with rescue of phenotype in the mouse model. Interestingly, the observed induction of TH was compromised on silencing of MTA1. The underlying molecular mechanism of MTA1 induction by G-CSF was proved to be through induction of c-Fos and its recruitment to the MTA1 promoter.
Collapse
Affiliation(s)
- Arathy S Kumar
- From the Department of Biotechnology, Indian Institute of Technology Madras (IITM), Chennai 600 036 and
| | - Sankar Jagadeeshan
- From the Department of Biotechnology, Indian Institute of Technology Madras (IITM), Chennai 600 036 and
| | - Anirudh Subramanian
- From the Department of Biotechnology, Indian Institute of Technology Madras (IITM), Chennai 600 036 and
| | | | | | - Mahak Singhal
- From the Department of Biotechnology, Indian Institute of Technology Madras (IITM), Chennai 600 036 and
| | | | - Sathiya Sekar
- the Centre for Toxicology and Developmental Research (CEFT)
| | | | - Prathiba Duvuru
- Pathology, Sri Ramachandra University, Porur, Chennai 600 116, India
| | | | - Suresh K Rayala
- From the Department of Biotechnology, Indian Institute of Technology Madras (IITM), Chennai 600 036 and
| |
Collapse
|
20
|
Asakawa T, Fang H, Sugiyama K, Nozaki T, Hong Z, Yang Y, Hua F, Ding G, Chao D, Fenoy AJ, Villarreal SJ, Onoe H, Suzuki K, Mori N, Namba H, Xia Y. Animal behavioral assessments in current research of Parkinson's disease. Neurosci Biobehav Rev 2016; 65:63-94. [PMID: 27026638 DOI: 10.1016/j.neubiorev.2016.03.016] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 03/22/2016] [Accepted: 03/22/2016] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD), a neurodegenerative disorder, is traditionally classified as a movement disorder. Patients typically suffer from many motor dysfunctions. Presently, clinicians and scientists recognize that many non-motor symptoms are associated with PD. There is an increasing interest in both motor and non-motor symptoms in clinical studies on PD patients and laboratory research on animal models that imitate the pathophysiologic features and symptoms of PD patients. Therefore, appropriate behavioral assessments are extremely crucial for correctly understanding the mechanisms of PD and accurately evaluating the efficacy and safety of novel therapies. This article systematically reviews the behavioral assessments, for both motor and non-motor symptoms, in various animal models involved in current PD research. We addressed the strengths and weaknesses of these behavioral tests and their appropriate applications. Moreover, we discussed potential mechanisms behind these behavioral tests and cautioned readers against potential experimental bias. Since most of the behavioral assessments currently used for non-motor symptoms are not particularly designed for animals with PD, it is of the utmost importance to greatly improve experimental design and evaluation in PD research with animal models. Indeed, it is essential to develop specific assessments for non-motor symptoms in PD animals based on their characteristics. We concluded with a prospective view for behavioral assessments with real-time assessment with mobile internet and wearable device in future PD research.
Collapse
Affiliation(s)
- Tetsuya Asakawa
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka, Japan; Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka, Japan.
| | - Huan Fang
- Department of Pharmacy, Jinshan Hospital of Fudan University, Shanghai, China
| | - Kenji Sugiyama
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka, Japan
| | - Takao Nozaki
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka, Japan
| | - Zhen Hong
- Department of Neurology, Huashan Hospital of Fudan University, Shanghai, China
| | - Yilin Yang
- The First People's Hospital of Changzhou, Soochow University School of Medicine, Changzhou, China
| | - Fei Hua
- The First People's Hospital of Changzhou, Soochow University School of Medicine, Changzhou, China
| | - Guanghong Ding
- Shanghai Key Laboratory of Acupuncture Mechanism and Acupoint Function, Fudan University, Shanghai, China
| | - Dongman Chao
- Department of Neurosurgery, The University of Texas McGovern Medical School,Houston, TX, USA
| | - Albert J Fenoy
- Department of Neurosurgery, The University of Texas McGovern Medical School,Houston, TX, USA
| | - Sebastian J Villarreal
- Department of Neurosurgery, The University of Texas McGovern Medical School,Houston, TX, USA
| | - Hirotaka Onoe
- Functional Probe Research Laboratory, RIKEN Center for Life Science Technologies, Kobe, Japan
| | - Katsuaki Suzuki
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka, Japan
| | - Norio Mori
- Department of Psychiatry, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka, Japan
| | - Hiroki Namba
- Department of Neurosurgery, Hamamatsu University School of Medicine, Hamamatsu-city, Shizuoka, Japan
| | - Ying Xia
- Department of Neurosurgery, The University of Texas McGovern Medical School,Houston, TX, USA.
| |
Collapse
|
21
|
Banks WA, Dohi K, Hansen K, Thompson HJ. Assessing blood granulocyte colony-stimulating factor as a potential biomarker of acute traumatic brain injury in mice and humans. Brain Behav Immun 2016; 52:81-87. [PMID: 26441136 PMCID: PMC5873950 DOI: 10.1016/j.bbi.2015.10.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 09/03/2015] [Accepted: 10/02/2015] [Indexed: 02/01/2023] Open
Abstract
Previous work has found that serum G-CSF was acutely elevated in mice 24h but not one week after controlled cortical impact (CCI). The purpose of this study was to investigate whether blood G-CSF correlates with the elevated brain cytokines in mice after CCI and also if it correlates with traumatic brain injury (TBI) in humans. Here, we found in mice undergoing CCI, a procedure that induces direct injury to the brain, that serum G-CSF correlated directly or indirectly with several brain cytokines, indicating it is a useful marker for the neuroinflammation of TBI. A pilot study in humans (phase I, n=19) confirmed that plasma G-CSF is acutely elevated on day 1 (p<0.001) of TBI and has returned to baseline by one week. In a second human sample (phase II) (n=80), we found plasma G-CSF peaks about 12h after arriving in the emergency department (41.6+/-5.4 pg/ml). Aging was weakly associated (p<0.05) with a less robust elevation in serum G-CSF, but there was no difference with gender. ISS, a measure of total severity of injury, correlated with the degree of elevation in serum G-CSF (r=.419; p<0.05), but severity of head injury (via AIS) did not. The latter may have been because of the statistically narrow range of head injuries among our cases and the high number of cases diagnosed with closed head injury (a non-codable diagnosis). In conclusion, plasma G-CSF may be a useful biomarker of TBI, correlating with neuroinflammation in the animal model and in the human studies with time since injury and total severity of injury. As such, it may be useful in determining whether TBI has occurred within the last 24h.
Collapse
Affiliation(s)
- William A. Banks
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA,Corresponding author at: Rm 1/810A, VAPSHCS, 1660 S. Columbian Way, Seattle, WA 98108, USA
| | - Kenji Dohi
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA; Department of Emergency Medicine, The Jikei University School of Medicine, Tokyo, Japan.
| | - Kim Hansen
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98104, USA.
| | - Hilaire J. Thompson
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA,Department of Biobehavioral Nursing & Health Systems, School of Nursing, University of Washington, Seattle, WA 98195, USA,Harborview Injury Prevention and Research Center, University of Washington, Seattle, WA 98104, USA
| |
Collapse
|
22
|
Banks WA. From blood-brain barrier to blood-brain interface: new opportunities for CNS drug delivery. Nat Rev Drug Discov 2016; 15:275-92. [PMID: 26794270 DOI: 10.1038/nrd.2015.21] [Citation(s) in RCA: 758] [Impact Index Per Article: 84.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
One of the biggest challenges in the development of therapeutics for central nervous system (CNS) disorders is achieving sufficient blood-brain barrier (BBB) penetration. Research in the past few decades has revealed that the BBB is not only a substantial barrier for drug delivery to the CNS but also a complex, dynamic interface that adapts to the needs of the CNS, responds to physiological changes, and is affected by and can even promote disease. This complexity confounds simple strategies for drug delivery to the CNS, but provides a wealth of opportunities and approaches for drug development. Here, I review some of the most important areas that have recently redefined the BBB and discuss how they can be applied to the development of CNS therapeutics.
Collapse
Affiliation(s)
- William A Banks
- Veterans Affairs Puget Sound Health Care System, Geriatrics Research Education and Clinical Center and Department of Medicine, University of Washington School of Medicine, Division of Gerontology and Geriatric Medicine, 1660 South Columbian Way, Seattle, Washington 98108, USA
| |
Collapse
|
23
|
Liu J, Huang D, Xu J, Tong J, Wang Z, Huang L, Yang Y, Bai X, Wang P, Suo H, Ma Y, Yu M, Fei J, Huang F. Tiagabine Protects Dopaminergic Neurons against Neurotoxins by Inhibiting Microglial Activation. Sci Rep 2015; 5:15720. [PMID: 26499517 PMCID: PMC4620555 DOI: 10.1038/srep15720] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 10/02/2015] [Indexed: 01/04/2023] Open
Abstract
Microglial activation and inflammation are associated with progressive neuronal apoptosis in neurodegenerative disorders such as Parkinson's disease (PD). γ-Aminobutyric acid (GABA), the major inhibitory neurotransmitter in the central nervous system, has recently been shown to play an inhibitory role in the immune system. Tiagabine, a piperidine derivative, enhances GABAergic transmission by inhibiting GABA transporter 1 (GAT 1). In the present study, we found that tiagabine pretreatment attenuated microglial activation, provided partial protection to the nigrostriatal axis and improved motor deficits in a methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. The protective function of tiagabine was abolished in GAT 1 knockout mice that were challenged with MPTP. In an alternative PD model, induced by intranigral infusion of lipopolysaccharide (LPS), microglial suppression and subsequent neuroprotective effects of tiagabine were demonstrated. Furthermore, the LPS-induced inflammatory activation of BV-2 microglial cells and the toxicity of conditioned medium toward SH-SY5Y cells were inhibited by pretreatment with GABAergic drugs. The attenuation of the nuclear translocation of nuclear factor κB (NF-κB) and the inhibition of the generation of inflammatory mediators were the underlying mechanisms. Our results suggest that tiagabine acts as a brake for nigrostriatal microglial activation and that it might be a novel therapeutic approach for PD.
Collapse
Affiliation(s)
- Jie Liu
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China.,Research Center for Translational Medicine and Institute of Heart Failure, East Hospital, Tongji University, Shanghai 200120, China
| | - Dongping Huang
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Jing Xu
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Jiabin Tong
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Zishan Wang
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Li Huang
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Yufang Yang
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Xiaochen Bai
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Pan Wang
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Haiyun Suo
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Yuanyuan Ma
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Mei Yu
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Jian Fei
- School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China.,Shanghai Research Center for Model Organisms, Pudong, Shanghai 201203, China
| | - Fang Huang
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China.,Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Shanghai 201203, China
| |
Collapse
|
24
|
Heinzelman P, Schoborg JA, Jewett MC. pH responsive granulocyte colony-stimulating factor variants with implications for treating Alzheimer's disease and other central nervous system disorders. Protein Eng Des Sel 2015; 28:481-9. [PMID: 25877663 PMCID: PMC4596278 DOI: 10.1093/protein/gzv022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 02/27/2015] [Accepted: 03/06/2015] [Indexed: 12/14/2022] Open
Abstract
Systemic injection of granulocyte colony-stimulating factor (G-CSF) has yielded encouraging results in treating Alzheimer's Disease (AD) and other central nervous system (CNS) disorders. Making G-CSF a viable AD therapeutic will, however, require increasing G-CSF's ability to stimulate neurons within the brain. This objective could be realized by increasing transcytosis of G-CSF across the blood brain barrier (BBB). An established correlation between G-CSF receptor (G-CSFR) binding pH responsiveness and increased recycling of G-CSF to the cell exterior after endocytosis motivated development of G-CSF variants with highly pH responsive G-CSFR binding affinities. These variants will be used in future validation of our hypothesis that increased BBB transcytosis can enhance G-CSF therapeutic efficacy. Flow cytometric screening of a yeast-displayed library in which G-CSF/G-CSFR interface residues were mutated to histidine yielded a G-CSF triple His mutant (L109H/D110H/Q120H) with highly pH responsive binding affinity. This variant's KD, measured by surface plasmon resonance (SPR), increases ∼20-fold as pH decreases from 7.4 to below histidine's pKa of ∼6.0; an increase 2-fold greater than for previously reported G-CSF His mutants. Cell-free protein synthesis (CFPS) enabled expression and purification of soluble, bioactive G-CSF triple His variant protein, an outcome inaccessible via Escherichia coli inclusion body refolding. This purification and bioactivity validation will enable future identification of correlations between pH responsiveness and transcytosis in BBB cell culture model and animal experiments. Furthermore, the library screening and CFPS methods employed here could be applied to developing other pH responsive hematopoietic or neurotrophic factors for treating CNS disorders.
Collapse
Affiliation(s)
- Pete Heinzelman
- Department of Chemical, Biological and Materials Engineering, University of Oklahoma, Sarkeys Energy Center, 100 East Boyd Street, Room T-301, Norman, OK 73019, USA
| | - Jennifer A Schoborg
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208-3120, USA
| | - Michael C Jewett
- Department of Chemical and Biological Engineering, Northwestern University, Evanston, IL 60208-3120, USA
| |
Collapse
|
25
|
The Role of Neutrophils in Measles Virus-mediated Oncolysis Differs Between B-cell Malignancies and Is Not Always Enhanced by GCSF. Mol Ther 2015; 24:184-92. [PMID: 26278331 DOI: 10.1038/mt.2015.149] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 08/10/2015] [Indexed: 02/08/2023] Open
Abstract
The mechanism by which oncolytic measles virus (MV) kills cancer cells remains obscure. We previously showed that neutrophils are involved in MV-mediated tumor regressions and become activated, upon MV infection. In the present study, we attempted to enhance the neutrophil response toward MV-infected tumor targets by generating an oncolytic MV-expressing human granulocyte colony-stimulating factor (MVhGCSF). Evaluating the effects in two different models of B-cell malignancy, we showed that depletion of neutrophils abrogated the MV therapeutic effect in an in vivo Raji-but not Nalm-6 tumor model. Next, we compared MVhGCSF with the unmodified backbone virus MVNSe. MVhGCSF enhanced the oncolytic capacity of MV in the Raji model in vivo, whereas in the Nalm-6 model, the opposite was unexpectedly the case. This finding was recapitulated by exogenously administered hGCSF. MVhGCSF replicated within an MV-infectable CD46 transgenic mouse model with detectable serum levels of hGCSF but no toxicity. Our data suggest that a "one-size-fits-all" model of immune response to viral oncolysis is not appropriate, and each tumor target will need full characterization for the potential of both direct and indirect, innate immune responses to generate benefit.
Collapse
|
26
|
Suo H, Wang P, Tong J, Cai L, Liu J, Huang D, Huang L, Wang Z, Huang Y, Xu J, Ma Y, Yu M, Fei J, Huang F. NRSF is an essential mediator for the neuroprotection of trichostatin A in the MPTP mouse model of Parkinson's disease. Neuropharmacology 2015; 99:67-78. [PMID: 26188143 DOI: 10.1016/j.neuropharm.2015.07.015] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Revised: 06/26/2015] [Accepted: 07/14/2015] [Indexed: 11/29/2022]
Abstract
Neuron-restrictive silencer factor (NRSF) blocks the expression of many neuronal genes in non-neuronal cells and neural stem cells. There is growing body of evidence that NRSF functions in mature neurons and plays critical roles in various neurological disorders. Our previous study demonstrated that the expression of NRSF target genes brain-derived neurotrophic factor (BDNF), and tyrosine hydroxylase (TH) is transiently decreased in 1-methyl-4-phenyl-pyridinium ion (MPP+)-treated SH-SY5Y cells. NRSF neuronal deficient mice are more vulnerable to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Here we investigated the effect of epigenetic modulation on the expression of NRSF target genes in in vitro and in vivo models of Parkinson's disease (PD). Trichostatin A (TSA) was further used to study the effects of histone deacetylase inhibition on NRSF-mediated repression. We found that the repression of NRSF target genes was relieved by TSA in vitro. A single dose TSA pretreatment also upregulated the expression of TH and BDNF and protected the nigrostriatal dopaminergic pathway against MPTP-induced degeneration in wild type mice. However, the protective functions of TSA were fully abolished in NRSF neuronal deficient mice. Our results suggest that NRSF serves as an essential mediator for the neuroprotection of TSA in the MPTP model of PD.
Collapse
Affiliation(s)
- Haiyun Suo
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Pan Wang
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Jiabin Tong
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Lei Cai
- Shanghai Research Center for Model Organisms, Pudong, Shanghai 201203, China
| | - Jie Liu
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Dongping Huang
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Li Huang
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Zishan Wang
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Yufang Huang
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Jing Xu
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Yuanyuan Ma
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China
| | - Mei Yu
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China.
| | - Jian Fei
- School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China; Shanghai Research Center for Model Organisms, Pudong, Shanghai 201203, China.
| | - Fang Huang
- The State Key Laboratory of Medical Neurobiology, the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Shanghai Medical College, Fudan University, 138 Yixueyuan Road, Shanghai 200032, China; Key Laboratory of Smart Drug Delivery, Fudan University, Ministry of Education, Shanghai 201203, China.
| |
Collapse
|
27
|
Gao L, Brenner D, Llorens-Bobadilla E, Saiz-Castro G, Frank T, Wieghofer P, Hill O, Thiemann M, Karray S, Prinz M, Weishaupt JH, Martin-Villalba A. Infiltration of circulating myeloid cells through CD95L contributes to neurodegeneration in mice. ACTA ACUST UNITED AC 2015; 212:469-80. [PMID: 25779632 PMCID: PMC4387281 DOI: 10.1084/jem.20132423] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 02/26/2015] [Indexed: 11/17/2022]
Abstract
Gao et al. report that genetic or pharmacological blockade of CD95 ligand prevents infiltration of peripheral myeloid cells and thereby averts toxin-induced neurodegeneration in mice. Neuroinflammation is increasingly recognized as a hallmark of neurodegeneration. Activated central nervous system–resident microglia and infiltrating immune cells contribute to the degeneration of dopaminergic neurons (DNs). However, how the inflammatory process leads to neuron loss and whether blocking this response would be beneficial to disease progression remains largely unknown. CD95 is a mediator of inflammation that has also been proposed as an apoptosis inducer in DNs, but previous studies using ubiquitous deletion of CD95 or CD95L in mouse models of neurodegeneration have generated conflicting results. Here we examine the role of CD95 in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridin (MPTP)–induced neurodegeneration using tissue-specific deletion of CD95 or CD95L. We show that DN death is not mediated by CD95-induced apoptosis because deletion of CD95 in DNs does not influence MPTP-induced neurodegeneration. In contrast, deletion of CD95L in peripheral myeloid cells significantly protects against MPTP neurotoxicity and preserves striatal dopamine levels. Systemic pharmacological inhibition of CD95L dampens the peripheral innate response, reduces the accumulation of infiltrating myeloid cells, and efficiently prevents MPTP-induced DN death. Altogether, this study emphasizes the role of the peripheral innate immune response in neurodegeneration and identifies CD95 as potential pharmacological target for neurodegenerative disease.
Collapse
Affiliation(s)
- Liang Gao
- Division of Molecular Neurobiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - David Brenner
- Division of Molecular Neurobiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Enric Llorens-Bobadilla
- Division of Molecular Neurobiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Gonzalo Saiz-Castro
- Division of Molecular Neurobiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Tobias Frank
- Department of Neurology, University Medical Center, Georg-August-University, 37075 Göttingen, Germany
| | - Peter Wieghofer
- Institute of Neuropathology, Faculty of Biology, and BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79098 Freiburg, Germany Institute of Neuropathology, Faculty of Biology, and BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79098 Freiburg, Germany
| | | | | | - Saoussen Karray
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1160, Université Paris Diderot, 75010 Paris, France
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Biology, and BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79098 Freiburg, Germany Institute of Neuropathology, Faculty of Biology, and BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79098 Freiburg, Germany
| | | | - Ana Martin-Villalba
- Division of Molecular Neurobiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
28
|
Kallmünzer B, Tauchi M, Schlachetzki JC, Machold K, Schmidt A, Winkler J, Schwab S, Kollmar R. Granulocyte colony-stimulating factor does not promote neurogenesis after experimental intracerebral haemorrhage. Int J Stroke 2013; 9:783-8. [PMID: 24920160 DOI: 10.1111/ijs.12217] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Hematopoietic growth factors have been suggested to induce neuroprotective and regenerative effects in various animal models of cerebral injury. However, the pathways involved remain widely unexplored. AIMS This study aimed to investigate effects of local and systemic administration of granulocyte colony-stimulating factor on brain damage, functional recovery, and cerebral neurogenesis in an intracerebral haemorrhage whole blood injection model in rats. METHODS Eight-week-old male Wistar rats (n = 100) underwent induction of striatal intracerebral haemorrhage by autologous whole blood injection or sham procedure and were randomly assigned to either (a) systemic treatment with granulocyte colony-stimulating factor (60 μg/kg) for five-days; (b) single intracerebral injection of granulocyte colony-stimulating factor (60 μg/kg) into the cavity; or (c) application of vehicle for five-days. Bromodeoxyuridine-labelling and immunohistochemistry were used to analyze proliferation and survival of newly born cells in the sub-ventricular zone and the hippocampal dentate gyrus. Moreover, functional deficits and lesion volume were assessed until day 42 after intracerebral haemorrhage. RESULTS Differences in lesion size or hemispheric atrophy between granulocyte colony-stimulating factor-treated and control groups did not reach statistical significance. Neither systemic, nor local granulocyte colony-stimulating factor administration induced neurogenesis within the dentate gyrus or the sub-ventricular zone. The survival of newborn cells in these regions was prevented by intracerebral granulocyte colony-stimulating factor application. A subtle benefit in functional recovery at day 14 after intracerebral haemorrhage induction was observed after granulocyte colony-stimulating factor treatment. CONCLUSION There was a lack of neuroprotective or neuroregenerative effects of granulocyte colony-stimulating factor in the present rodent model of intracerebral haemorrhage. Conflicting results from functional outcome assessment require further research.
Collapse
Affiliation(s)
- Bernd Kallmünzer
- Department of Neurology, University Medical Center Erlangen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
29
|
G-CSF attenuates noise-induced hearing loss. Neurosci Lett 2013; 562:102-6. [PMID: 23916659 DOI: 10.1016/j.neulet.2013.07.033] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 07/20/2013] [Accepted: 07/20/2013] [Indexed: 02/04/2023]
Abstract
In this study, we investigated the effects of granulocyte colony-stimulating factor (G-CSF) for the treatment of noise-induced hearing loss (NIHL) in a guinea pig model. Forty guinea pigs were randomly divided into four groups: control, noise (white noise, 3 h/d for 2 days at 115 dB), noise+G-CSF (350 μg/kg/d for 5 days), and noise+saline. Auditory brainstem response (ABR) and distortion product otoacoustic emission (DPOAE) were used to determine the hearing threshold and outer hair cell function, respectively, in each group. Cochlear morphology was examined to evaluate hair cell injury induced by intense noise exposure. Fourteen days after noise exposure, the noise+G-CSF group had a lower ABR value than the noise group (P<0.05) or the noise+saline group (P<0.01). At most frequencies, the DPOAE value of the noise+G-CSF group showed a significant rise (P<0.05) compared to the noise group or the noise+saline group. Neither the ABR value nor the DPOAE value differed between the noise group and the noise+saline group. The morphology of the phalloidin-stained organ of Corti was consistent with the functional measurements. In conclusion, G-CSF can preserve hearing in an experimental model of NIHL in guinea pigs, by preserving hair cells after intense noise exposure.
Collapse
|
30
|
Tönges L, Frank T, Tatenhorst L, Saal KA, Koch JC, Szegő ÉM, Bähr M, Weishaupt JH, Lingor P. Inhibition of rho kinase enhances survival of dopaminergic neurons and attenuates axonal loss in a mouse model of Parkinson's disease. Brain 2012; 135:3355-70. [PMID: 23087045 PMCID: PMC3501973 DOI: 10.1093/brain/aws254] [Citation(s) in RCA: 136] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 07/18/2012] [Accepted: 07/21/2012] [Indexed: 01/08/2023] Open
Abstract
Axonal degeneration is one of the earliest features of Parkinson's disease pathology, which is followed by neuronal death in the substantia nigra and other parts of the brain. Inhibition of axonal degeneration combined with cellular neuroprotection therefore seem key to targeting an early stage in Parkinson's disease progression. Based on our previous studies in traumatic and neurodegenerative disease models, we have identified rho kinase as a molecular target that can be manipulated to disinhibit axonal regeneration and improve survival of lesioned central nervous system neurons. In this study, we examined the neuroprotective potential of pharmacological rho kinase inhibition mediated by fasudil in the in vitro 1-methyl-4-phenylpyridinium cell culture model and in the subchronic in vivo 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson's disease. Application of fasudil resulted in a significant attenuation of dopaminergic cell loss in both paradigms. Furthermore, dopaminergic terminals were preserved as demonstrated by analysis of neurite network in vitro, striatal fibre density and by neurochemical analysis of the levels of dopamine and its metabolites in the striatum. Behavioural tests demonstrated a clear improvement in motor performance after fasudil treatment. The Akt survival pathway was identified as an important molecular mediator for neuroprotective effects of rho kinase inhibition in our paradigm. We conclude that inhibition of rho kinase using the clinically approved small molecule inhibitor fasudil may be a promising new therapeutic strategy for Parkinson's disease.
Collapse
MESH Headings
- 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives
- 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology
- 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/therapeutic use
- 1-Methyl-4-phenylpyridinium/toxicity
- Animals
- Axons/drug effects
- Axons/pathology
- Behavior, Animal/drug effects
- Behavior, Animal/physiology
- Cell Survival/drug effects
- Cell Survival/physiology
- Cells, Cultured
- Corpus Striatum/metabolism
- Disease Models, Animal
- Dopamine/metabolism
- Dopaminergic Neurons/enzymology
- Dopaminergic Neurons/pathology
- Dopaminergic Neurons/physiology
- MPTP Poisoning/drug therapy
- MPTP Poisoning/enzymology
- Male
- Mice
- Mice, Inbred C57BL
- Nerve Degeneration/chemically induced
- Nerve Degeneration/drug therapy
- Nerve Degeneration/enzymology
- Neurites/pathology
- Neuroprotective Agents/metabolism
- Neuroprotective Agents/pharmacology
- Neuroprotective Agents/therapeutic use
- Parkinson Disease, Secondary/chemically induced
- Parkinson Disease, Secondary/drug therapy
- Parkinson Disease, Secondary/enzymology
- Parkinson Disease, Secondary/pathology
- Proto-Oncogene Proteins c-akt/drug effects
- Proto-Oncogene Proteins c-akt/metabolism
- Rats
- Rats, Wistar
- Substantia Nigra/drug effects
- Substantia Nigra/enzymology
- rho-Associated Kinases/antagonists & inhibitors
- rho-Associated Kinases/physiology
Collapse
Affiliation(s)
- Lars Tönges
- 1 Department of Neurology, University Medicine Göttingen, 37075 Göttingen, Germany
| | - Tobias Frank
- 1 Department of Neurology, University Medicine Göttingen, 37075 Göttingen, Germany
| | - Lars Tatenhorst
- 1 Department of Neurology, University Medicine Göttingen, 37075 Göttingen, Germany
| | - Kim A. Saal
- 1 Department of Neurology, University Medicine Göttingen, 37075 Göttingen, Germany
| | - Jan C. Koch
- 1 Department of Neurology, University Medicine Göttingen, 37075 Göttingen, Germany
| | - Éva M. Szegő
- 2 Cluster of Excellence “Nanoscale Microscopy and Molecular Physiology of the Brain” (CNMPB), 37075 Göttingen, Germany
- 3 Department of Neurodegeneration and Restorative Research, University of Göttingen, 37075 Göttingen, Germany
| | - Mathias Bähr
- 1 Department of Neurology, University Medicine Göttingen, 37075 Göttingen, Germany
- 2 Cluster of Excellence “Nanoscale Microscopy and Molecular Physiology of the Brain” (CNMPB), 37075 Göttingen, Germany
| | | | - Paul Lingor
- 1 Department of Neurology, University Medicine Göttingen, 37075 Göttingen, Germany
- 2 Cluster of Excellence “Nanoscale Microscopy and Molecular Physiology of the Brain” (CNMPB), 37075 Göttingen, Germany
| |
Collapse
|