101
|
Valekova I, Jarkovska K, Kotrcova E, Bucci J, Ellederova Z, Juhas S, Motlik J, Gadher SJ, Kovarova H. Revelation of the IFNα, IL-10, IL-8 and IL-1β as promising biomarkers reflecting immuno-pathological mechanisms in porcine Huntington's disease model. J Neuroimmunol 2016; 293:71-81. [PMID: 27049565 DOI: 10.1016/j.jneuroim.2016.02.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 02/12/2016] [Accepted: 02/17/2016] [Indexed: 12/25/2022]
Abstract
Studies on Huntington's disease (HD) demonstrated altered immune response in HD gene carriers. Using multiplexing immunoassay, we simultaneously investigated seven cytokines in secretomes of microglia and blood monocytes, cerebrospinal fluid (CSF) and serum collected from transgenic HD minipigs at pre-symptomatic disease stage. Decline in IFNα and IL-10 was observed in CSF and secretome of microglia whilst elevated IL-8 and IL-1β levels were secreted by microglia. Additionally, IL-8 was increased in serum. The proportion of mutant huntingtin in microglia may have causative impact on cytokine production. IFNα, IL-10, IL-8 and IL-1β represent promising biomarkers reflecting immuno-pathological mechanisms in porcine HD model.
Collapse
Affiliation(s)
- Ivona Valekova
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, v.v.i., Rumburska 89, 277 21 Libechov, Czech Republic; Research Center PIGMOD, Rumburska 89, 277 21 Libechov, Czech Republic; Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, 128 43 Prague, Czech Republic
| | - Karla Jarkovska
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, v.v.i., Rumburska 89, 277 21 Libechov, Czech Republic; Research Center PIGMOD, Rumburska 89, 277 21 Libechov, Czech Republic
| | - Eva Kotrcova
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, v.v.i., Rumburska 89, 277 21 Libechov, Czech Republic; Research Center PIGMOD, Rumburska 89, 277 21 Libechov, Czech Republic; Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, 128 43 Prague, Czech Republic
| | - John Bucci
- Thermo Fisher Scientific, 5791 Van Allen Way, Carlsbad, CA 92008, USA
| | - Zdenka Ellederova
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, v.v.i., Rumburska 89, 277 21 Libechov, Czech Republic; Research Center PIGMOD, Rumburska 89, 277 21 Libechov, Czech Republic
| | - Stefan Juhas
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, v.v.i., Rumburska 89, 277 21 Libechov, Czech Republic; Research Center PIGMOD, Rumburska 89, 277 21 Libechov, Czech Republic
| | - Jan Motlik
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, v.v.i., Rumburska 89, 277 21 Libechov, Czech Republic; Research Center PIGMOD, Rumburska 89, 277 21 Libechov, Czech Republic
| | - Suresh Jivan Gadher
- Thermo Fisher Scientific - Life Science Solutions, 7335 Executive Way, Frederick, MD 21704, USA
| | - Hana Kovarova
- Institute of Animal Physiology and Genetics, Academy of Sciences of the Czech Republic, v.v.i., Rumburska 89, 277 21 Libechov, Czech Republic; Research Center PIGMOD, Rumburska 89, 277 21 Libechov, Czech Republic.
| |
Collapse
|
102
|
Recombinant Adeno Associated Viral (AAV) vector type 9 delivery of Ex1-Q138-mutant huntingtin in the rat striatum as a short-time model for in vivo studies in drug discovery. Neurobiol Dis 2016; 86:41-51. [DOI: 10.1016/j.nbd.2015.11.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 09/30/2015] [Accepted: 11/23/2015] [Indexed: 11/18/2022] Open
|
103
|
Fisher SP, Schwartz MD, Wurts-Black S, Thomas AM, Chen TM, Miller MA, Palmerston JB, Kilduff TS, Morairty SR. Quantitative Electroencephalographic Analysis Provides an Early-Stage Indicator of Disease Onset and Progression in the zQ175 Knock-In Mouse Model of Huntington's Disease. Sleep 2016; 39:379-91. [PMID: 26446107 DOI: 10.5665/sleep.5448] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 09/05/2015] [Indexed: 01/09/2023] Open
Abstract
STUDY OBJECTIVES Patients with Huntington's disease (HD) show a high prevalence of sleep disorders that typically occur prior to the onset of motoric symptoms and neurodegeneration. Our understanding of the pathophysiological alterations in premanifest HD is limited, hindering the ability to measure disease modification in response to treatment. We used a full-length knock-in HD model to determine early changes in the electroencephalogram (EEG) and sleep that may predict the onset and progression of the disease. METHODS A 10-month longitudinal study was designed to determine the effect of the HD mutation on the EEG and sleep/wake changes in heterozygous (HET) and homozygous (HOM) zQ175 mice and wild-type (WT) littermates from 8 to 48 w of age. Mice were instrumented with tethered headmounts to record EEG/electromyography signals. Telemeters were implanted to continuously measure locomotor activity (LMA) and body temperature (Tb). Sleep deprivation (SDep) was performed at 8, 12, 16, 24, 32, and 48 w of age. RESULTS The HD mutation disrupted the EEG field potential from 8-12 w in an age- and mutant huntington dose-dependent manner, prior to changes in sleep/wake states, LMA, and Tb. Prominent effects of the HD mutation on the EEG included a progressive reduction in low frequency power, a slowing of rapid eye movement peak theta frequency, and the emergence of state-dependent beta/gamma oscillations. There was no effect of genotype on the relative increase in nonrapid eye movement delta power or sleep time in response to SDep. CONCLUSIONS The expression of the Huntington's disease (HD) mutation results in complex EEG alterations that occur prior to deficits in behavioral measures and are one of the earliest phenotypes uncovered in this mouse model. Despite these EEG changes, homeostatic responses to sleep loss were preserved in HET and HOM zQ175 mice. Greater insight into the localization and response of these EEG alterations to novel therapies may enable early intervention and improve outcomes for patients with HD.
Collapse
Affiliation(s)
- Simon P Fisher
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California
| | - Michael D Schwartz
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California
| | - Sarah Wurts-Black
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California
| | - Alexia M Thomas
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California
| | - Tsui-Ming Chen
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California
| | - Michael A Miller
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California
| | - Jeremiah B Palmerston
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California
| | - Thomas S Kilduff
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California
| | - Stephen R Morairty
- Center for Neuroscience, Biosciences Division, SRI International, Menlo Park, California
| |
Collapse
|
104
|
Li P, Nie Y, Yu J. An Effective Method to Identify Shared Pathways and Common Factors among Neurodegenerative Diseases. PLoS One 2015; 10:e0143045. [PMID: 26575483 PMCID: PMC4648499 DOI: 10.1371/journal.pone.0143045] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 10/29/2015] [Indexed: 11/18/2022] Open
Abstract
Groups of distinct but related diseases often share common symptoms, which suggest likely overlaps in underlying pathogenic mechanisms. Identifying the shared pathways and common factors among those disorders can be expected to deepen our understanding for them and help designing new treatment strategies effected on those diseases. Neurodegeneration diseases, including Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD), were taken as a case study in this research. Reported susceptibility genes for AD, PD and HD were collected and human protein-protein interaction network (hPPIN) was used to identify biological pathways related to neurodegeneration. 81 KEGG pathways were found to be correlated with neurodegenerative disorders. 36 out of the 81 are human disease pathways, and the remaining ones are involved in miscellaneous human functional pathways. Cancers and infectious diseases are two major subclasses within the disease group. Apoptosis is one of the most significant functional pathways. Most of those pathways found here are actually consistent with prior knowledge of neurodegenerative diseases except two cell communication pathways: adherens and tight junctions. Gene expression analysis showed a high probability that the two pathways were related to neurodegenerative diseases. A combination of common susceptibility genes and hPPIN is an effective method to study shared pathways involved in a group of closely related disorders. Common modules, which might play a bridging role in linking neurodegenerative disorders and the enriched pathways, were identified by clustering analysis. The identified shared pathways and common modules can be expected to yield clues for effective target discovery efforts on neurodegeneration.
Collapse
Affiliation(s)
- Ping Li
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yaling Nie
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jingkai Yu
- National Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- * E-mail:
| |
Collapse
|
105
|
Politis M, Lahiri N, Niccolini F, Su P, Wu K, Giannetti P, Scahill RI, Turkheimer FE, Tabrizi SJ, Piccini P. Increased central microglial activation associated with peripheral cytokine levels in premanifest Huntington's disease gene carriers. Neurobiol Dis 2015; 83:115-21. [PMID: 26297319 DOI: 10.1016/j.nbd.2015.08.011] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 07/19/2015] [Accepted: 08/12/2015] [Indexed: 01/27/2023] Open
Abstract
Previous studies have shown activation of the immune system and altered immune response in Huntington's disease (HD) gene carriers. Here, we hypothesized that peripheral and central immune responses could be concurrent pathophysiological events and represent a global innate immune response to the toxic effects of mutant huntingtin in HD gene carriers. We sought to investigate our hypothesis using [(11)C]PK11195 PET as a translocator protein (TSPO) marker of central microglial activation, together with assessment of peripheral plasma cytokine levels in a cohort of premanifest HD gene carriers who were more than a decade from predicted symptomatic conversion. Data were also compared to those from a group of healthy controls matched for age and gender. We found significantly increased peripheral plasma IL-1β levels in premanifest HD gene carriers compared to the group of normal controls (P=0.018). Premanifest HD gene carriers had increased TSPO levels in cortical, basal ganglia and thalamic brain regions (P<0.001). Increased microglial activation in somatosensory cortex correlated with higher plasma levels of IL-1β (rs=0.87, P=0.013), IL-6 (rs=0.85, P=0.013), IL-8 (rs=0.68, P=0.045) and TNF-α (rs=0.79; P=0.013). Our findings provide first in vivo evidence for an association between peripheral and central immune responses in premanifest HD gene carriers, and provide further supporting evidence for the role of immune dysfunction in the pathogenesis of HD.
Collapse
Affiliation(s)
- Marios Politis
- Neurodegeneration Imaging Group, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - Nayana Lahiri
- Huntington's Disease Research Group, Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, UK
| | - Flavia Niccolini
- Neurodegeneration Imaging Group, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - Paul Su
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - Kit Wu
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - Paolo Giannetti
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - Rachael I Scahill
- Huntington's Disease Research Group, Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, UK
| | - Federico E Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Sarah J Tabrizi
- Huntington's Disease Research Group, Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, UK
| | - Paola Piccini
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK.
| |
Collapse
|
106
|
Andre R, Carty L, Tabrizi SJ. Disruption of immune cell function by mutant huntingtin in Huntington's disease pathogenesis. Curr Opin Pharmacol 2015; 26:33-8. [PMID: 26461267 DOI: 10.1016/j.coph.2015.09.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 09/15/2015] [Accepted: 09/16/2015] [Indexed: 12/28/2022]
Abstract
Innate immune dysfunction is increasingly recognised as a key characteristic of neurodegenerative disease. In the fatal inherited neurological disorder, Huntington's disease, altered innate immune cell function and increased inflammation are observed in the brain and the periphery of disease gene carriers many years before symptom onset, suggesting a potentially early and important role in disease pathogenesis. This is due, at least in part, to the intrinsic effects of the disease-causing protein, mutant huntingtin, expressed in innate immune cells themselves. Understanding whether such innate immune dysfunction in Huntington's disease can be targeted to slow the onset and/or the progression of the disease has significant therapeutic implications and is the subject of much current research.
Collapse
Affiliation(s)
- Ralph Andre
- UCL Institute of Neurology, Department of Neurodegenerative Disease, London, UK
| | - Lucy Carty
- UCL Institute of Neurology, Department of Neurodegenerative Disease, London, UK
| | - Sarah J Tabrizi
- UCL Institute of Neurology, Department of Neurodegenerative Disease, London, UK.
| |
Collapse
|
107
|
Niccolini F, Haider S, Reis Marques T, Muhlert N, Tziortzi AC, Searle GE, Natesan S, Piccini P, Kapur S, Rabiner EA, Gunn RN, Tabrizi SJ, Politis M. Altered PDE10A expression detectable early before symptomatic onset in Huntington's disease. Brain 2015; 138:3016-29. [PMID: 26198591 DOI: 10.1093/brain/awv214] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 05/28/2015] [Indexed: 12/28/2022] Open
Abstract
There is an urgent need for early biomarkers and novel disease-modifying therapies in Huntington's disease. Huntington's disease pathology involves the toxic effect of mutant huntingtin primarily in striatal medium spiny neurons, which highly express phosphodiesterase 10A (PDE10A). PDE10A hydrolyses cAMP/cGMP signalling cascades, thus having a key role in the regulation of striatal output, and in promoting neuronal survival. PDE10A could be a key therapeutic target in Huntington's disease. Here, we used combined positron emission tomography (PET) and multimodal magnetic resonance imaging to assess PDE10A expression in vivo in a unique cohort of 12 early premanifest Huntington's disease gene carriers with a mean estimated 90% probability of 25 years before the predicted onset of clinical symptoms. We show bidirectional changes in PDE10A expression in premanifest Huntington's disease gene carriers, which are associated with the probability of symptomatic onset. PDE10A expression in early premanifest Huntington's disease was decreased in striatum and pallidum and increased in motor thalamic nuclei, compared to a group of matched healthy controls. Connectivity-based analysis revealed prominent PDE10A decreases confined in the sensorimotor-striatum and in striatonigral and striatopallidal projecting segments. The ratio between higher PDE10A expression in motor thalamic nuclei and lower PDE10A expression in striatopallidal projecting striatum was the strongest correlate with higher probability of symptomatic conversion in early premanifest Huntington's disease gene carriers. Our findings demonstrate in vivo, a novel and earliest pathophysiological mechanism underlying Huntington's disease with direct implications for the development of new pharmacological treatments, which can promote neuronal survival and improve outcome in Huntington's disease gene carriers.
Collapse
Affiliation(s)
- Flavia Niccolini
- 1 Neurodegeneration Imaging Group, Department of Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK 2 Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - Salman Haider
- 3 Huntington's Disease Research Group, Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, UK
| | - Tiago Reis Marques
- 4 Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Nils Muhlert
- 5 School of Psychology and Cardiff University Brain Research Imaging Centre, Cardiff University, UK 6 School of Psychological Sciences, University of Manchester, Manchester, UK
| | - Andri C Tziortzi
- 7 Imanova Ltd., Centre for Imaging Sciences, Hammersmith Hospital, London, UK
| | - Graham E Searle
- 7 Imanova Ltd., Centre for Imaging Sciences, Hammersmith Hospital, London, UK
| | - Sridhar Natesan
- 4 Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Paola Piccini
- 2 Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - Shitij Kapur
- 4 Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Eugenii A Rabiner
- 7 Imanova Ltd., Centre for Imaging Sciences, Hammersmith Hospital, London, UK 8 Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King s College London, London, UK
| | - Roger N Gunn
- 2 Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK 7 Imanova Ltd., Centre for Imaging Sciences, Hammersmith Hospital, London, UK
| | - Sarah J Tabrizi
- 3 Huntington's Disease Research Group, Department of Neurodegenerative Disease, Institute of Neurology, University College London, London, UK
| | - Marios Politis
- 1 Neurodegeneration Imaging Group, Department of Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK 2 Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| |
Collapse
|
108
|
Wang HL, Ma RH, Fang H, Xue ZG, Liao QW. Impaired Spatial Learning Memory after Isoflurane Anesthesia or Appendectomy in Aged Mice is Associated with Microglia Activation. J Cell Death 2015; 8:9-19. [PMID: 26380557 PMCID: PMC4560456 DOI: 10.4137/jcd.s30596] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 08/03/2015] [Accepted: 08/06/2015] [Indexed: 12/18/2022] Open
Abstract
Postoperative cognitive dysfunction (POCD) has been one of the most common problems in elderly patients following surgery. But the specific mechanism of POCD is still not clear. To further understand the reason of these postoperative behavioral deficits, we evaluated the spatial learning memory of both adult (3 months) and aged (18 months) male mice, 3 or 28 days after isoflurane (Iso) exposure for two hours or appendectomy (App). Hippocampal microglia activation and IL-1β, TNF-α, and IFN-γ expression were also evaluated at day 3, day 14 and day 28 after Iso exposure or appendectomy. Results showed that spatial learning memory of aged, but not adult, mice was impaired after Iso exposure or appendectomy, accompanied with more hippocampal microglia activation and IL-1β, TNF-α, and IFN-γ overexpression. These findings suggest that the cognitive deficits of elderly patients who have undergone surgeries are quite possibly caused by hippocampal microglia overactivation and the subsequent inflammation.
Collapse
Affiliation(s)
- Hui-Lin Wang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Rui-Hua Ma
- Department of Anesthesiology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Hao Fang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China. ; Department of Anesthesiology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Zhang-Gang Xue
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qing-Wu Liao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
109
|
Alakurtti K, Johansson JJ, Joutsa J, Laine M, Bäckman L, Nyberg L, Rinne JO. Long-term test-retest reliability of striatal and extrastriatal dopamine D2/3 receptor binding: study with [(11)C]raclopride and high-resolution PET. J Cereb Blood Flow Metab 2015; 35:1199-205. [PMID: 25853904 PMCID: PMC4640276 DOI: 10.1038/jcbfm.2015.53] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 02/27/2015] [Accepted: 03/02/2015] [Indexed: 01/13/2023]
Abstract
We measured the long-term test-retest reliability of [(11)C]raclopride binding in striatal subregions, the thalamus and the cortex using the bolus-plus-infusion method and a high-resolution positron emission scanner. Seven healthy male volunteers underwent two positron emission tomography (PET) [(11)C]raclopride assessments, with a 5-week retest interval. D2/3 receptor availability was quantified as binding potential using the simplified reference tissue model. Absolute variability (VAR) and intraclass correlation coefficient (ICC) values indicated very good reproducibility for the striatum and were 4.5%/0.82, 3.9%/0.83, and 3.9%/0.82, for the caudate nucleus, putamen, and ventral striatum, respectively. Thalamic reliability was also very good, with VAR of 3.7% and ICC of 0.92. Test-retest data for cortical areas showed good to moderate reproducibility (6.1% to 13.1%). Our results are in line with previous test-retest studies of [(11)C]raclopride binding in the striatum. A novel finding is the relatively low variability of [(11)C]raclopride binding, providing suggestive evidence that extrastriatal D2/3 binding can be studied in vivo with [(11)C]raclopride PET to be verified in future studies.
Collapse
Affiliation(s)
- Kati Alakurtti
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Jarkko J Johansson
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Juho Joutsa
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Matti Laine
- Department of Psychology and Logopedics, Abo Akademi University, Turku, Finland
| | - Lars Bäckman
- Aging Research Center, Karolinska Institute, Stockholm, Sweden
| | - Lars Nyberg
- Department of Radiation Sciences, Umeå University, Umeå, Sweden
| | - Juha O Rinne
- 1] Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland [2] Department of Neurology, Turku University Hospital, Turku, Finland
| |
Collapse
|
110
|
Mattner F, Quinlivan M, Greguric I, Pham T, Liu X, Jackson T, Berghofer P, Fookes CJR, Dikic B, Gregoire MC, Dolle F, Katsifis A. Radiosynthesis, In Vivo Biological Evaluation, and Imaging of Brain Lesions with [123I]-CLINME, a New SPECT Tracer for the Translocator Protein. DISEASE MARKERS 2015; 2015:729698. [PMID: 26199457 PMCID: PMC4496498 DOI: 10.1155/2015/729698] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 06/10/2015] [Indexed: 11/17/2022]
Abstract
The high affinity translocator protein (TSPO) ligand 6-chloro-2-(4'-iodophenyl)-3-(N,N-methylethyl)imidazo[1,2-a]pyridine-3-acetamide (CLINME) was radiolabelled with iodine-123 and assessed for its sensitivity for the TSPO in rodents. Moreover neuroinflammatory changes on a unilateral excitotoxic lesion rat model were detected using SPECT imaging. [(123)I]-CLINME was prepared in 70-80% radiochemical yield. The uptake of [(123)I]-CLINME was evaluated in rats by biodistribution, competition, and metabolite studies. The unilateral excitotoxic lesion was performed by injection of α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid unilaterally into the striatum. The striatum lesion was confirmed and correlated with TSPO expression in astrocytes and activated microglia by immunohistochemistry and autoradiography. In vivo studies with [(123)I]-CLINME indicated a biodistribution pattern consistent with TPSO distribution and the competition studies with PK11195 and Ro 5-4864 showed that [(123)I]-CLINME is selective for this site. The metabolite study showed that the extractable radioactivity was unchanged [(123)I]-CLINME in organs which expresses TSPO. SPECT/CT imaging on the unilateral excitotoxic lesion indicated that the mean ratio uptake in striatum (lesion:nonlesion) was 2.2. Moreover, TSPO changes observed by SPECT imaging were confirmed by immunofluorescence, immunochemistry, and autoradiography. These results indicated that [(123)I]-CLINME is a promising candidate for the quantification and visualization of TPSO expression in activated astroglia using SPECT.
Collapse
Affiliation(s)
- F. Mattner
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
- Department of Molecular Imaging, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - M. Quinlivan
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - I. Greguric
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - T. Pham
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - X. Liu
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - T. Jackson
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - P. Berghofer
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - C. J. R. Fookes
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - B. Dikic
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - M.-C. Gregoire
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
| | - F. Dolle
- CEA, DSV/I2BM, Service Hospitalier Frédéric Joliot, 4 Place du Général Leclerc, 91401 Orsay, France
| | - A. Katsifis
- Life Sciences Division, Australian Nuclear Science and Technology Organisation, New Illawarra Road, Lucas Heights, NSW 2234, Australia
- Department of Molecular Imaging, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| |
Collapse
|
111
|
Lin C, Lin HY, Chen JH, Tseng WP, Ko PY, Liu YS, Yeh WL, Lu DY. Effects of paeonol on anti-neuroinflammatory responses in microglial cells. Int J Mol Sci 2015; 16:8844-60. [PMID: 25906473 PMCID: PMC4425112 DOI: 10.3390/ijms16048844] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 04/07/2015] [Accepted: 04/14/2015] [Indexed: 12/17/2022] Open
Abstract
Increasing studies suggest that inflammatory processes in the central nervous system mediated by microglial activation plays an important role in numerous neurodegenerative diseases. Development of planning for microglial suppression is considered a key strategy in the search for neuroprotection. Paeonol is a major phenolic component of Moutan Cortex, widely used as a nutrient supplement in Chinese medicine. In this study, we investigated the effects of paeonol on microglial cells stimulated by inflammagens. Paeonol significantly inhibited the release of nitric oxide (NO) and the expressions of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Treatment with paeonol also reduced reactive oxygen species (ROS) production and inhibited an ATP-induced increased cell migratory activity. Furthermore, the inhibitory effects of neuroinflammation by paeonol were found to be regulated by phosphorylated adenosine monophosphate-activated protein kinase-α (AMPK-α) and glycogen synthase kinase 3 α/β (GSK 3α/β). Treatment with AMPK or GSK3 inhibitors reverse the inhibitory effect of neuroinflammation by paeonol in microglial cells. Furthermore, paeonol treatment also showed significant improvement in the rotarod performance and microglial activation in the mouse model as well. The present study is the first to report a novel inhibitory role of paeonol on neuroinflammation, and presents a new candidate agent for the development of therapies for inflammation-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Chingju Lin
- Department of Physiology, School of Medicine, China Medical University, Taichung 40402, Taiwan.
| | - Hsiao-Yun Lin
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, Taichung 40402, Taiwan.
| | - Jia-Hong Chen
- Department of General Surgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung 42743, Taiwan.
| | - Wen-Pei Tseng
- Graduate Institute of Sports and Health, National Changhua University of Education, Changhua 500, Taiwan.
| | - Pei-Ying Ko
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 40402, Taiwan.
| | - Yu-Shu Liu
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan.
| | - Wei-Lan Yeh
- Department of Cell and Tissue Engineering, Changhua Christian Hospital, Changhua 500, Taiwan.
| | - Dah-Yuu Lu
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, Taichung 40402, Taiwan.
- Department of Photonics and Communication Engineering, Asia University, Taichung 40402, Taiwan.
| |
Collapse
|
112
|
Recent imaging advances in neurology. J Neurol 2015; 262:2182-94. [PMID: 25808503 DOI: 10.1007/s00415-015-7711-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 03/13/2015] [Accepted: 03/14/2015] [Indexed: 01/08/2023]
Abstract
Over the recent years, the application of neuroimaging techniques such as magnetic resonance imaging (MRI) and positron emission tomography (PET) has considerably advanced the understanding of complex neurological disorders. PET is a powerful molecular imaging tool, which investigates the distribution and binding of radiochemicals attached to biologically relevant molecules; as such, this technique is able to give information on biochemistry and metabolism of the brain in health and disease. MRI uses high intensity magnetic fields and radiofrequency pulses to provide structural and functional information on tissues and organs in intact or diseased individuals, including the evaluation of white matter integrity, grey matter thickness and brain perfusion. The aim of this article is to review the most recent advances in neuroimaging research in common neurological disorders such as movement disorders, dementia, epilepsy, traumatic brain injury and multiple sclerosis, and to evaluate their contribution in the diagnosis and management of patients.
Collapse
|
113
|
Tsai CF, Kuo YH, Yeh WL, Wu CYJ, Lin HY, Lai SW, Liu YS, Wu LH, Lu JK, Lu DY. Regulatory effects of caffeic acid phenethyl ester on neuroinflammation in microglial cells. Int J Mol Sci 2015; 16:5572-89. [PMID: 25768341 PMCID: PMC4394493 DOI: 10.3390/ijms16035572] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Revised: 02/12/2015] [Accepted: 02/28/2015] [Indexed: 12/20/2022] Open
Abstract
Microglial activation has been widely demonstrated to mediate inflammatory processes that are crucial in several neurodegenerative disorders. Pharmaceuticals that can deliver direct inhibitory effects on microglia are therefore considered as a potential strategy to counter balance neurodegenerative progression. Caffeic acid phenethyl ester (CAPE), a natural phenol in honeybee propolis, is known to possess antioxidant, anti-inflammatory and anti-microbial properties. Accordingly, the current study intended to probe the effects of CAPE on microglia activation by using in vitro and in vivo models. Western blot and Griess reaction assay revealed CAPE significantly inhibited the expressions of inducible nitric oxide synthase (NOS), cyclooxygenase (COX)-2 and the production of nitric oxide (NO). Administration of CAPE resulted in increased expressions of hemeoxygenase (HO)-1and erythropoietin (EPO) in microglia. The phosphorylated adenosine monophosphate-activated protein kinase (AMPK)-α was further found to regulate the anti-inflammatory effects of caffeic acid. In vivo results from immunohistochemistry along with rotarod test also revealed the anti-neuroinflammatory effects of CAPE in microglia activation. The current study has evidenced several possible molecular determinants, AMPKα, EPO, and HO-1, in mediating anti-neuroinflammatory responses in microglial cells.
Collapse
Affiliation(s)
- Cheng-Fang Tsai
- Department of Biotechnology, Asia University, Taichung 413, Taiwan.
| | - Yueh-Hsiung Kuo
- Department of Biotechnology, Asia University, Taichung 413, Taiwan.
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, China Medical University, Taichung 404, Taiwan.
| | - Wei-Lan Yeh
- Department of Cell and Tissue Engineering, Changhua Christian Hospital, Changhua 500, Taiwan.
| | - Caren Yu-Ju Wu
- Graduate Institute of Basic Medical Science, College of Medicine, China Medical University, Taichung 404, Taiwan.
| | - Hsiao-Yun Lin
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, Taichung 404, Taiwan.
| | - Sheng-Wei Lai
- Graduate Institute of Basic Medical Science, College of Medicine, China Medical University, Taichung 404, Taiwan.
| | - Yu-Shu Liu
- Graduate Institute of Basic Medical Science, College of Medicine, China Medical University, Taichung 404, Taiwan.
| | - Ling-Hsuan Wu
- Graduate Institute of Basic Medical Science, College of Medicine, China Medical University, Taichung 404, Taiwan.
| | - Jheng-Kun Lu
- Department of Biotechnology, Asia University, Taichung 413, Taiwan.
| | - Dah-Yuu Lu
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, Taichung 404, Taiwan.
- Department of Photonics and Communication Engineering, Asia University, Taichung 413, Taiwan.
| |
Collapse
|
114
|
Süssmuth SD, Haider S, Landwehrmeyer GB, Farmer R, Frost C, Tripepi G, Andersen CA, Di Bacco M, Lamanna C, Diodato E, Massai L, Diamanti D, Mori E, Magnoni L, Dreyhaupt J, Schiefele K, Craufurd D, Saft C, Rudzinska M, Ryglewicz D, Orth M, Brzozy S, Baran A, Pollio G, Andre R, Tabrizi SJ, Darpo B, Westerberg G. An exploratory double-blind, randomized clinical trial with selisistat, a SirT1 inhibitor, in patients with Huntington's disease. Br J Clin Pharmacol 2015; 79:465-76. [PMID: 25223731 PMCID: PMC4345957 DOI: 10.1111/bcp.12512] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Accepted: 09/06/2014] [Indexed: 12/22/2022] Open
Abstract
AIMS Selisistat, a selective SirT1 inhibitor is being developed as a potentially disease-modifying therapeutic for Huntington's disease (HD). This was the first study of selisistat in HD patients and was primarily aimed at development of pharmacodynamic biomarkers. METHODS This was a randomized, double-blind, placebo-controlled, multicentre exploratory study. Fifty-five male and female patients in early stage HD were randomized to receive 10 mg or 100 mg of selisistat or placebo once daily for 14 days. Blood sampling, clinical and safety assessments were conducted throughout the study. Candidate pharmacodynamic markers included circulating soluble huntingtin and innate immune markers. RESULTS Selisistat was found to be safe and well tolerated, and systemic exposure parameters showed that the average steady-state plasma concentration achieved at the 10 mg dose level (125 nm) was comparable with the IC50 for SirT1 inhibition. No adverse effects on motor, cognitive or functional readouts were recorded. While circulating levels of soluble huntingtin were not affected by selisistat in this study, the biological samples collected have allowed development of assay technology for use in future studies. No effects on innate immune markers were seen. CONCLUSIONS Selisistat was found to be safe and well tolerated in early stage HD patients at plasma concentrations within the anticipated therapeutic concentration range.
Collapse
Affiliation(s)
| | - Salman Haider
- Department of Neurodegenerative Disease, University College London Institute of NeurologyLondon, United Kingdom
| | | | - Ruth Farmer
- Department of Medical Statistics, London School of Hygiene and Tropical MedicineLondon, United Kingdom
| | - Chris Frost
- Department of Medical Statistics, London School of Hygiene and Tropical MedicineLondon, United Kingdom
| | | | | | | | - Claudia Lamanna
- Siena Biotech SpASiena, Italy
- European Huntington's Disease Network (EHDN)Chieti, Italy
| | | | | | | | | | | | - Jens Dreyhaupt
- Institute of Epidemiology and Medical Biometry, Ulm UniversityUlm, Germany
| | - Karin Schiefele
- Institute of Epidemiology and Medical Biometry, Ulm UniversityUlm, Germany
| | - David Craufurd
- Institute of Human Development, Faculty of Medical and Human Sciences, University of ManchesterManchester, United Kingdom
- Manchester Centre for Genomic Medicine, Central Manchester University Hospitals NHS Foundation Trust and Manchester Academic Health Science CentreManchester, United Kingdom
| | - Carsten Saft
- Department of Neurology, Huntington-Center NRW, St. Josef-Hospital, Ruhr-UniversityBochum, Germany
| | - Monika Rudzinska
- Department of Neurology, Medical University of SilesiaKatowice, Poland
| | - Danuta Ryglewicz
- Department of Neurology, Institute of Psychiatry and NeurologyWarsaw, Poland
| | - Michael Orth
- Department of Neurology, Ulm University HospitalUlm, Germany
| | | | | | | | - Ralph Andre
- Department of Neurodegenerative Disease, University College London Institute of NeurologyLondon, United Kingdom
| | - Sarah J Tabrizi
- Department of Neurodegenerative Disease, University College London Institute of NeurologyLondon, United Kingdom
| | - Borje Darpo
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Karolinska Institutet, Danderyd HospitalStockholm, Sweden
- iCardiac TechnologiesRochester, NY, USA
| | - Goran Westerberg
- Siena Biotech SpASiena, Italy
- La Crocina Pharmaceutical Consultants D.I.San Giovanni d'Asso (S), Italy
| | - Paddington Consortium
- Department of Neurology, Ulm University HospitalUlm, Germany
- Department of Neurodegenerative Disease, University College London Institute of NeurologyLondon, United Kingdom
- Department of Medical Statistics, London School of Hygiene and Tropical MedicineLondon, United Kingdom
- Siena Biotech SpASiena, Italy
- European Huntington's Disease Network (EHDN)Chieti, Italy
- Institute of Epidemiology and Medical Biometry, Ulm UniversityUlm, Germany
- Institute of Human Development, Faculty of Medical and Human Sciences, University of ManchesterManchester, United Kingdom
- Manchester Centre for Genomic Medicine, Central Manchester University Hospitals NHS Foundation Trust and Manchester Academic Health Science CentreManchester, United Kingdom
- Department of Neurology, Huntington-Center NRW, St. Josef-Hospital, Ruhr-UniversityBochum, Germany
- Department of Neurology, Medical University of SilesiaKatowice, Poland
- Department of Neurology, Institute of Psychiatry and NeurologyWarsaw, Poland
- KCR.S.A.Warsaw, Poland
- Division of Cardiovascular Medicine, Department of Clinical Sciences, Karolinska Institutet, Danderyd HospitalStockholm, Sweden
- iCardiac TechnologiesRochester, NY, USA
- La Crocina Pharmaceutical Consultants D.I.San Giovanni d'Asso (S), Italy
| |
Collapse
|
115
|
Gabery S, Georgiou-Karistianis N, Lundh SH, Cheong RY, Churchyard A, Chua P, Stout JC, Egan GF, Kirik D, Petersén Å. Volumetric analysis of the hypothalamus in Huntington Disease using 3T MRI: the IMAGE-HD Study. PLoS One 2015; 10:e0117593. [PMID: 25659157 PMCID: PMC4319930 DOI: 10.1371/journal.pone.0117593] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 12/29/2014] [Indexed: 12/25/2022] Open
Abstract
Huntington disease (HD) is a fatal neurodegenerative disorder caused by an expanded CAG repeat in the huntingtin gene. Non-motor symptoms and signs such as psychiatric disturbances, sleep problems and metabolic dysfunction are part of the disease manifestation. These aspects may relate to changes in the hypothalamus, an area of the brain involved in the regulation of emotion, sleep and metabolism. Neuropathological and imaging studies using both voxel-based morphometry (VBM) of magnetic resonance imaging (MRI) as well as positron emission tomography (PET) have demonstrated pathological changes in the hypothalamic region during early stages in symptomatic HD. In this investigation, we aimed to establish a robust method for measurements of the hypothalamic volume in MRI in order to determine whether the hypothalamic dysfunction in HD is associated with the volume of this region. Using T1-weighted imaging, we describe a reproducible delineation procedure to estimate the hypothalamic volume which was based on the same landmarks used in histologically processed postmortem hypothalamic tissue. Participants included 36 prodromal HD (pre-HD), 33 symptomatic HD (symp-HD) and 33 control participants who underwent MRI scanning at baseline and 18 months follow-up as part of the IMAGE-HD study. We found no evidence of cross-sectional or longitudinal changes between groups in hypothalamic volume. Our results suggest that hypothalamic pathology in HD is not associated with volume changes.
Collapse
Affiliation(s)
- Sanaz Gabery
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Sofia Hult Lundh
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Rachel Y. Cheong
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Andrew Churchyard
- School of Psychological Sciences, Monash University, Clayton, Victoria, 3180, Australia
- Huntington’s Disease Unit, Bethlehem Hospital, Kooyong Rd, Caulfield, Victoria, 3162, Australia
| | - Phyllis Chua
- Huntington’s Disease Unit, Bethlehem Hospital, Kooyong Rd, Caulfield, Victoria, 3162, Australia
- Department of Psychiatry, School of Clinical Sciences at Monash Health, Monash University, Clayton, Victoria, 3168, Australia
| | - Julie C. Stout
- School of Psychological Sciences, Monash University, Clayton, Victoria, 3180, Australia
| | - Gary F. Egan
- School of Psychological Sciences, Monash University, Clayton, Victoria, 3180, Australia
- Monash Biomedical Imaging (MBI), Monash University, Clayton, Victoria, 3180, Australia
- Life Sciences Computation Centre, Victorian Life Sciences Computation Initiative (VLSCI), Melbourne, Victoria, Australia
| | - Deniz Kirik
- Brain Repair and Imaging in Neural Systems (B.R.A.I.N.S) Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Lund University Bioimaging Center, Lund, Sweden
| | - Åsa Petersén
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
116
|
Prodromal Huntington disease as a model for functional compensation of early neurodegeneration. PLoS One 2014; 9:e114569. [PMID: 25541992 PMCID: PMC4277279 DOI: 10.1371/journal.pone.0114569] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 11/12/2014] [Indexed: 11/19/2022] Open
Abstract
Functional compensation demonstrated as mechanism to offset neuronal loss in early Alzheimer disease may also occur in other adult-onset neurodegenerative diseases, particularly Huntington disease (HD) with its genetic determination and gradual changes in structural integrity. In HD, neurodegeneration typically initiates in the dorsal striatum, successively affecting ventral striatal areas. Investigating carriers of the HD mutation with evident dorsal, but only minimal or no ventral striatal atrophy, we expected to find evidence for compensation of ventral striatal functioning. We investigated 14 pre- or early symptomatic carriers of the mutation leading to HD and 18 matched healthy controls. Participants underwent structural T1 magnetic resonance imaging (MRI) and functional MRI during a reward task that probes ventral striatal functioning. Motor functioning and attention were assessed with reaction time (RT) tasks. Structural images confirmed a specific decrease of dorsal striatal but only marginal ventral striatal volume in HD relative to control subjects, paralleling prolonged RT in the motor response tasks. While behavioral performance in the reward task during fMRI scanning was unimpaired, reward-related fMRI signaling in the HD group was differentially enhanced in the bilateral ventral striatum and in bilateral orbitofrontal cortex/anterior insula, as another region sensitive to reward processing. We provide evidence for the concept of functional compensation in premanifest HD which may suggest a defense mechanism in neurodegeneration. Given the so far inevitable course of HD with its genetically determined endpoint, this disease may provide another model to study the different aspects of the concept of functional compensation.
Collapse
|
117
|
Yuan Y, Zha H, Rangarajan P, Ling EA, Wu C. Anti-inflammatory effects of Edaravone and Scutellarin in activated microglia in experimentally induced ischemia injury in rats and in BV-2 microglia. BMC Neurosci 2014; 15:125. [PMID: 25416145 PMCID: PMC4247200 DOI: 10.1186/s12868-014-0125-3] [Citation(s) in RCA: 91] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 11/10/2014] [Indexed: 01/13/2023] Open
Abstract
Background In response to cerebral ischemia, activated microglia release excessive inflammatory mediators which contribute to neuronal damage. Therefore, inhibition of microglial over-activation could be a therapeutic strategy to alleviate various microglia-mediated neuroinflammation. This study was aimed to elucidate the anti-inflammatory effects of Scutellarin and Edaravone given either singly, or in combination in activated microglia in rats subjected to middle cerebral artery occlusion (MCAO), and in lipopolysaccharide (LPS)-induced BV-2 microglia. Expression of proinflammatory cytokines, including tumor necrosis factor-alpha (TNF-α), interleukin-1 beta (IL-1β), and inducible nitric oxide synthase (iNOS) was assessed by immunofluorescence staining and Western blot. Reactive oxygen species (ROS) and nitric oxide (NO) levels were determined by flow cytometry and fluorescence microscopy, respectively. Results In vivo, both Edaravone and Scutellarin markedly reduced the infarct cerebral tissue area with the latter drug being more effective with the dosage used; furthermore, when used in combination the reduction was more substantial. Remarkably, a greater diminution in distribution of activated microglia was observed with the combined drug treatment which also attenuated the immunoexpression of TNF-α, IL-1β and iNOS to a greater extent as compared to the drugs given separately. In vitro, both drugs suppressed upregulated expression of inflammatory cytokines, iNOS, NO and ROS in LPS-induced BV-2 cells. Furthermore, Edaravone and Scutellarin in combination cumulatively diminished the expression levels of the inflammatory mediators being most pronounced for TNF-α as evidenced by Western blot. Conclusion The results suggest that Edaravone and Scutellarin effectively suppressed the inflammatory responses in activated microglia, with Scutellarin being more efficacious within the dosage range used. Moreover, when both drugs were used in combination, the infarct tissue area was reduced more extensively; also, microglia-mediated inflammatory mediators notably TNF-α expression was decreased cumulatively. Electronic supplementary material The online version of this article (doi:10.1186/s12868-014-0125-3) contains supplementary material, which is available to authorized users.
Collapse
|
118
|
Imaging neuroinflammation in Alzheimer's disease and other dementias: Recent advances and future directions. Alzheimers Dement 2014; 11:1110-20. [DOI: 10.1016/j.jalz.2014.08.105] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 04/21/2014] [Accepted: 08/12/2014] [Indexed: 12/13/2022]
|
119
|
Abstract
Huntington’s disease (HD) is a progressive and fatal neurodegenerative disorder caused by an expanded trinucleotide CAG sequence in huntingtin gene (HTT) on chromosome 4. HD manifests with chorea, cognitive and psychiatric symptoms. Although advances in genetics allow identification of individuals carrying the HD gene, much is still unknown about the mechanisms underlying the development of overt clinical symptoms and the transitional period between premanifestation and manifestation of the disease. HD has no cure and patients rely only in symptomatic treatment. There is an urgent need to identify biomarkers that are able to monitor disease progression and assess the development and efficacy of novel disease modifying drugs. Over the past years, neuroimaging techniques such as magnetic resonance imaging (MRI) and positron emission tomography (PET) have provided important advances in our understanding of HD. MRI provides information about structural and functional organization of the brain, while PET can detect molecular changes in the brain. MRI and PET are able to detect changes in the brains of HD gene carriers years ahead of the manifestation of the disease and have also proved to be powerful in assessing disease progression. However, no single technique has been validated as an optimal biomarker. An integrative multimodal imaging approach, which combines different MRI and PET techniques, could be recommended for monitoring potential neuroprotective and preventive therapies in HD. In this article we review the current neuroimaging literature in HD.
Collapse
|
120
|
Beste C, Mückschel M, Elben S, J Hartmann C, McIntyre CC, Saft C, Vesper J, Schnitzler A, Wojtecki L. Behavioral and neurophysiological evidence for the enhancement of cognitive control under dorsal pallidal deep brain stimulation in Huntington's disease. Brain Struct Funct 2014; 220:2441-8. [PMID: 24878825 DOI: 10.1007/s00429-014-0805-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 05/19/2014] [Indexed: 12/18/2022]
Abstract
Deep brain stimulation of the dorsal pallidum (globus pallidus, GP) is increasingly considered as a surgical therapeutic option in Huntington's disease (HD), but there is need to identify outcome measures useful for clinical trials. Computational models consider the GP to be part of a basal ganglia network involved in cognitive processes related to the control of actions. We examined behavioural and event-related potential (ERP) correlates of action control (i.e., error monitoring) and evaluated the effects of deep brain stimulation (DBS). We did this using a standard flanker paradigm and evaluated error-related ERPs. Patients were recruited from a prospective pilot trial for pallidal DBS in HD (trial number NCT00902889). From the initial four patients with Huntington's chorea, two patients with chronic external dorsal pallidum stimulation were available for follow-up and able to perform the task. The results suggest that the external GP constitutes an important basal ganglia element not only for error processing and behavioural adaptation but for general response monitoring processes as well. Response monitoring functions were fully controllable by switching pallidal DBS stimulation on and off. When stimulation was switched off, no neurophysiological and behavioural signs of error and general performance monitoring, as reflected by the error-related negativity and post-error slowing in reaction times were evident. The modulation of response monitoring processes by GP-DBS reflects a side effect of efforts to alleviate motor symptoms in HD. From a clinical neurological perspective, the results suggest that DBS in the external GP segment can be regarded as a potentially beneficial treatment with respect to cognitive functions.
Collapse
Affiliation(s)
- Christian Beste
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine of the TU Dresden, Schubertstrasse 42, 01309, Dresden, Germany,
| | | | | | | | | | | | | | | | | |
Collapse
|
121
|
Italiani P, Carlesi C, Giungato P, Puxeddu I, Borroni B, Bossù P, Migliorini P, Siciliano G, Boraschi D. Evaluating the levels of interleukin-1 family cytokines in sporadic amyotrophic lateral sclerosis. J Neuroinflammation 2014; 11:94. [PMID: 24884937 PMCID: PMC4039322 DOI: 10.1186/1742-2094-11-94] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 05/05/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Amyotrophic lateral sclerosis (ALS) is a progressive motor neuron disease leading to the death of affected individuals within years. The involvement of inflammation in the pathogenesis of neurodegenerative diseases, including ALS, is increasingly recognized but still not well understood. The aim of this study is to evaluate the levels of inflammation-related IL-1 family cytokines (IL-1β, IL-18, IL-33, IL-37) and their endogenous inhibitors (IL-1Ra, sIL-1R2, IL-18BP, sIL-1R4) in patients with sporadic ALS (sALS), METHODS: Sera were collected from 144 patients (125 patients were characterized by disease form, duration, and disability, using the revised ALS functional rating scale (ALSFRS-R) and from 40 matched controls. Cerebrospinal fluid (CSF) was collected from 54 patients with sALS and 65 patients with other non-infectious non-oncogenic diseases as controls. Cytokines and inhibitors were measured by commercial ELISA. RESULTS Among the IL-1 family cytokines tested total IL-18, its endogenous inhibitor IL-18BP, and the active form of the cytokine (free IL-18) were significantly higher in the sALS sera than in controls. No correlation between these soluble mediators and different clinical forms of sALS or the clinical setting of the disease was found. IL-18BP was the only mediator detectable in the CSF of patients. CONCLUSIONS Among the IL-1 family cytokines, only IL-18 correlates with this disease and may therefore have a pathological role in sALS. The increase of total IL-18 suggests the activation of IL-18-cleaving inflammasome. Whether IL-18 upregulation in circulation of sALS patients is a consequence of inflammation or one of the causes of the pathology still needs to be addressed.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Diana Boraschi
- Laboratory of Innate Immunity and Cytokines, Institute of Biomedical Technologies, National Research Council, Via G, Moruzzi 1, 56124 Pisa, Italy.
| |
Collapse
|
122
|
Alto LT, Chen X, Ruhn KA, Treviño I, Tansey MG. AAV-dominant negative tumor necrosis factor (DN-TNF) gene transfer to the striatum does not rescue medium spiny neurons in the YAC128 mouse model of Huntington's disease. PLoS One 2014; 9:e96544. [PMID: 24824433 PMCID: PMC4019512 DOI: 10.1371/journal.pone.0096544] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 04/08/2014] [Indexed: 11/19/2022] Open
Abstract
CNS inflammation is a hallmark of neurodegenerative disease, and recent studies suggest that the inflammatory response may contribute to neuronal demise. In particular, increased tumor necrosis factor (TNF) signaling is implicated in the pathology of both Parkinson's disease (PD) and Alzheimer's disease (AD). We have previously shown that localized gene delivery of dominant negative TNF to the degenerating brain region can limit pathology in animal models of PD and AD. TNF is upregulated in Huntington's disease (HD), like in PD and AD, but it is unknown whether TNF signaling contributes to neuronal degeneration in HD. We used in vivo gene delivery to test whether selective reduction of soluble TNF signaling could attenuate medium spiny neuron (MSN) degeneration in the YAC128 transgenic (TG) mouse model of Huntington's disease (HD). AAV vectors encoding cDNA for dominant-negative tumor necrosis factor (DN-TNF) or GFP (control) were injected into the striatum of young adult wild type WT and YAC128 TG mice and achieved 30-50% target coverage. Expression of dominant negative TNF protein was confirmed immunohistologically and biochemically and was maintained as mice aged to one year, but declined significantly over time. However, the extent of striatal DN-TNF gene transfer achieved in our studies was not sufficient to achieve robust effects on neuroinflammation, rescue degenerating MSNs or improve motor function in treated mice. Our findings suggest that alternative drug delivery strategies should be explored to determine whether greater target coverage by DN-TNF protein might afford some level of neuroprotection against HD-like pathology and/or that soluble TNF signaling may not be the primary driver of striatal neuroinflammation and MSN loss in YAC128 TG mice.
Collapse
Affiliation(s)
- Laura Taylor Alto
- Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Xi Chen
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Kelly A. Ruhn
- Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Isaac Treviño
- Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Malú G. Tansey
- Department of Physiology, The University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Department of Physiology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
123
|
Gomez-Nicola D, Perry VH. Microglial dynamics and role in the healthy and diseased brain: a paradigm of functional plasticity. Neuroscientist 2014; 21:169-84. [PMID: 24722525 PMCID: PMC4412879 DOI: 10.1177/1073858414530512] [Citation(s) in RCA: 235] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The study of the dynamics and functions of microglia in the healthy and diseased brain is a matter of intense scientific activity. The application of new techniques and new experimental approaches has allowed the identification of novel microglial functions and the redefinition of classic ones. In this review, we propose the study of microglial functions, rather than their molecular profiles, to better understand and define the roles of these cells in the brain. We review current knowledge on the role of surveillant microglia, proliferating microglia, pruning/neuromodulatory microglia, phagocytic microglia, and inflammatory microglia and the molecular profiles that are associated with these functions. In the remodeling scenario of microglial biology, the analysis of microglial functional states will inform about the roles in health and disease and will guide us to a more precise understanding of the multifaceted roles of this never-resting cells.
Collapse
Affiliation(s)
- Diego Gomez-Nicola
- Centre for Biological Sciences, University of Southampton, Southampton, UK
| | - V Hugh Perry
- Centre for Biological Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
124
|
Andre R, Scahill RI, Haider S, Tabrizi SJ. Biomarker development for Huntington's disease. Drug Discov Today 2014; 19:972-9. [PMID: 24632006 DOI: 10.1016/j.drudis.2014.03.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 03/04/2014] [Indexed: 02/02/2023]
Abstract
Huntington's disease (HD) is a fatal inherited neurodegenerative disorder, treatment to slow the progression of which has not yet been found. Human clinical trials to test a number of therapeutic strategies are underway or imminent, facilitated in part by the recent development of biomarkers that might be used as surrogate endpoints in such trials. However, although much progress in developing HD biomarkers has been made, ongoing work seeks to improve the sensitivity and reliability of current measures, and to demonstrate that they correspond to clear meaningful benefit to patients. Of particular importance is the identification of state biomarkers that can be used in pre-manifest HD gene carriers to test therapies hoped to delay symptom onset in these individuals. Functional, neuroimaging and biochemical biomarkers continue to be investigated for use in the development of disease-modifying treatments of HD.
Collapse
Affiliation(s)
- Ralph Andre
- UCL Institute of Neurology, Department of Neurodegenerative Disease, Queen Square, London WC1N 3BG, UK
| | - Rachael I Scahill
- UCL Institute of Neurology, Department of Neurodegenerative Disease, Queen Square, London WC1N 3BG, UK
| | - Salman Haider
- UCL Institute of Neurology, Department of Neurodegenerative Disease, Queen Square, London WC1N 3BG, UK
| | - Sarah J Tabrizi
- UCL Institute of Neurology, Department of Neurodegenerative Disease, Queen Square, London WC1N 3BG, UK.
| |
Collapse
|
125
|
Huang BR, Chang PC, Yeh WL, Lee CH, Tsai CF, Lin C, Lin HY, Liu YS, Wu CYJ, Ko PY, Huang SS, Hsu HC, Lu DY. Anti-neuroinflammatory effects of the calcium channel blocker nicardipine on microglial cells: implications for neuroprotection. PLoS One 2014; 9:e91167. [PMID: 24621589 PMCID: PMC3951295 DOI: 10.1371/journal.pone.0091167] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Accepted: 02/11/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/OBJECTIVE Nicardipine is a calcium channel blocker that has been widely used to control blood pressure in severe hypertension following events such as ischemic stroke, traumatic brain injury, and intracerebral hemorrhage. However, accumulating evidence suggests that inflammatory processes in the central nervous system that are mediated by microglial activation play important roles in neurodegeneration, and the effect of nicardipine on microglial activation remains unresolved. METHODOLOGY/PRINCIPAL FINDINGS In the present study, using murine BV-2 microglia, we demonstrated that nicardipine significantly inhibits microglia-related neuroinflammatory responses. Treatment with nicardipine inhibited microglial cell migration. Nicardipine also significantly inhibited LPS plus IFN-γ-induced release of nitric oxide (NO), and the expression of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2). Furthermore, nicardipine also inhibited microglial activation by peptidoglycan, the major component of the Gram-positive bacterium cell wall. Notably, nicardipine also showed significant anti-neuroinflammatory effects on microglial activation in mice in vivo. CONCLUSION/SIGNIFICANCE The present study is the first to report a novel inhibitory role of nicardipine on neuroinflammation and provides a new candidate agent for the development of therapies for inflammation-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Bor-Ren Huang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Neurosurgery Department, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Pei-Chun Chang
- Department of Bioinformatics, Asia University, Taichung, Taiwan
| | - Wei-Lan Yeh
- Department of Cell and Tissue Engineering, Changhua Christian Hospital, Changhua, Taiwan
| | - Chih-Hao Lee
- Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, United States of America
| | - Cheng-Fang Tsai
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Chingju Lin
- Department of Physiology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Hsiao-Yun Lin
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Yu-Shu Liu
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Caren Yu-Ju Wu
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Pei-Ying Ko
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan
| | - Shiang-Suo Huang
- Department of Pharmacology and Institute of Medicine, College of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Horng-Chaung Hsu
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Dah-Yuu Lu
- Graduate Institute of Neural and Cognitive Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
126
|
Crotti A, Benner C, Kerman BE, Gosselin D, Lagier-Tourenne C, Zuccato C, Cattaneo E, Gage FH, Cleveland DW, Glass CK. Mutant Huntingtin promotes autonomous microglia activation via myeloid lineage-determining factors. Nat Neurosci 2014; 17:513-21. [PMID: 24584051 DOI: 10.1038/nn.3668] [Citation(s) in RCA: 240] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 01/27/2014] [Indexed: 12/14/2022]
Abstract
Huntington's disease (HD) is a fatal neurodegenerative disorder caused by an extended polyglutamine repeat in the N terminus of the Huntingtin protein (HTT). Reactive microglia and elevated cytokine levels are observed in the brains of HD patients, but the extent to which neuroinflammation results from extrinsic or cell-autonomous mechanisms in microglia is unknown. Using genome-wide approaches, we found that expression of mutant Huntingtin (mHTT) in microglia promoted cell-autonomous pro-inflammatory transcriptional activation by increasing the expression and transcriptional activities of the myeloid lineage-determining factors PU.1 and C/EBPs. We observed elevated levels of PU.1 and its target genes in the brains of mouse models and individuals with HD. Moreover, mHTT-expressing microglia exhibited an increased capacity to induce neuronal death ex vivo and in vivo in the presence of sterile inflammation. These findings suggest a cell-autonomous basis for enhanced microglia reactivity that may influence non-cell-autonomous HD pathogenesis.
Collapse
Affiliation(s)
- Andrea Crotti
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, USA
| | - Christopher Benner
- Razavi Newman Integrative Genomics and Bioinformatics Core, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Bilal E Kerman
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, California, USA
| | - David Gosselin
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, USA
| | - Clotilde Lagier-Tourenne
- 1] Department of Neurosciences, University of California, San Diego, La Jolla, California, USA. [2] Ludwig Institute for Cancer Research, La Jolla, California, USA
| | - Chiara Zuccato
- Department of BioSciences and Center for Stem Cell Research, Università degli Studi di Milano, Milan, Italy
| | - Elena Cattaneo
- Department of BioSciences and Center for Stem Cell Research, Università degli Studi di Milano, Milan, Italy
| | - Fred H Gage
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, California, USA
| | - Don W Cleveland
- 1] Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, USA. [2] Ludwig Institute for Cancer Research, La Jolla, California, USA
| | - Christopher K Glass
- 1] Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, USA. [2] Department of Medicine, University of California, La Jolla, San Diego, California, USA
| |
Collapse
|
127
|
Yu M, Li X, Wu S, Shen J, Tu J. Examination of Huntington's disease in a Chinese family. Neural Regen Res 2014; 9:440-6. [PMID: 25206833 PMCID: PMC4146191 DOI: 10.4103/1673-5374.128258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2013] [Indexed: 01/01/2023] Open
Abstract
We report brain imaging and genetic diagnosis in a family from Wuhan, China, with a history of Huntington's disease. Among 17 family members across three generations, four patients (II2, II6, III5, and III9) show typical Huntington's disease, involuntary dance-like movements. Magnetic resonance imaging found lateral ventricular atrophy in three members (II2, II6, and III5). Moreover, genetic analysis identified abnormally amplified CAG sequence repeats (> 40) in two members (III5 and III9). Among borderline cases, with clinical symptoms and brain imaging features of Huntington's disease, two cases were identified (II2 and II6), but shown by mutation analysis for CAG expansions in the important transcript 15 gene, to be non-Huntington's disease. Our findings suggest that clinical diagnosis of Huntington's disease requires a combination of clinical symptoms, radiological changes, and genetic diagnosis.
Collapse
Affiliation(s)
- Mingxia Yu
- Department of Clinical Laboratory Medicine & Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Xiaogai Li
- Department of Clinical Laboratory Medicine & Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Sanyun Wu
- Department of Clinical Laboratory Medicine & Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Ji Shen
- Department of Clinical Laboratory Medicine & Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Jiancheng Tu
- Department of Clinical Laboratory Medicine & Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
128
|
Abstract
Neuroinflammation plays a central role in a variety of neurological diseases, including stroke, multiple sclerosis, Alzheimer’s disease, and malignant CNS neoplasms, among many other. Different cell types and molecular mediators participate in a cascade of events in the brain that is ultimately aimed at control, regeneration and repair, but leads to damage of brain tissue under pathological conditions. Non-invasive molecular imaging of key players in the inflammation cascade holds promise for identification and quantification of the disease process before it is too late for effective therapeutic intervention. In this review, we focus on molecular imaging techniques that target inflammatory cells and molecules that are of interest in neuroinflammation, especially those with high translational potential. Over the past decade, a plethora of molecular imaging agents have been developed and tested in animal models of (neuro)inflammation, and a few have been translated from bench to bedside. The most promising imaging techniques to visualize neuroinflammation include MRI, positron emission tomography (PET), single photon emission computed tomography (SPECT), and optical imaging methods. These techniques enable us to image adhesion molecules to visualize endothelial cell activation, assess leukocyte functions such as oxidative stress, granule release, and phagocytosis, and label a variety of inflammatory cells for cell tracking experiments. In addition, several cell types and their activation can be specifically targeted in vivo, and consequences of neuroinflammation such as neuronal death and demyelination can be quantified. As we continue to make progress in utilizing molecular imaging technology to study and understand neuroinflammation, increasing efforts and investment should be made to bring more of these novel imaging agents from the “bench to bedside.”
Collapse
Affiliation(s)
- Benjamin Pulli
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA 02114, USA
| | - John W Chen
- Center for Systems Biology and Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, MA 02114, USA
| |
Collapse
|
129
|
|
130
|
Hossain MM, Sonsalla PK, Richardson JR. Coordinated role of voltage-gated sodium channels and the Na+/H+ exchanger in sustaining microglial activation during inflammation. Toxicol Appl Pharmacol 2013; 273:355-64. [PMID: 24070585 DOI: 10.1016/j.taap.2013.09.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 08/23/2013] [Accepted: 09/13/2013] [Indexed: 11/18/2022]
Abstract
Persistent neuroinflammation and microglial activation play an integral role in the pathogenesis of many neurological disorders. We investigated the role of voltage-gated sodium channels (VGSC) and Na(+)/H(+) exchangers (NHE) in the activation of immortalized microglial cells (BV-2) after lipopolysaccharide (LPS) exposure. LPS (10 and 100 ng/ml) caused a dose- and time-dependent accumulation of intracellular sodium [(Na(+))i] in BV-2 cells. Pre-treatment of cells with the VGSC antagonist tetrodotoxin (TTX, 1 μM) abolished short-term Na(+) influx, but was unable to prevent the accumulation of (Na(+))i observed at 6 and 24h after LPS exposure. The NHE inhibitor cariporide (1 μM) significantly reduced accumulation of (Na(+))i 6 and 24h after LPS exposure. Furthermore, LPS increased the mRNA expression and protein level of NHE-1 in a dose- and time-dependent manner, which was significantly reduced after co-treatment with TTX and/or cariporide. LPS increased production of TNF-α, ROS, and H2O2 and expression of gp91(phox), an active subunit of NADPH oxidase, in a dose- and time-dependent manner, which was significantly reduced by TTX or TTX+cariporide. Collectively, these data demonstrate a closely-linked temporal relationship between VGSC and NHE-1 in regulating function in activated microglia, which may provide avenues for therapeutic interventions aimed at reducing neuroinflammation.
Collapse
Affiliation(s)
- Muhammad M Hossain
- Department of Environmental and Occupational Medicine and Environmental and Occupational Health Sciences Institute, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | | | | |
Collapse
|
131
|
Mutant huntingtin gene-dose impacts on aggregate deposition, DARPP32 expression and neuroinflammation in HdhQ150 mice. PLoS One 2013; 8:e75108. [PMID: 24086450 PMCID: PMC3781050 DOI: 10.1371/journal.pone.0075108] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 08/12/2013] [Indexed: 12/05/2022] Open
Abstract
Huntington's disease (HD) is an autosomal dominant, progressive and fatal neurological disorder caused by an expansion of CAG repeats in exon-1 of the huntingtin gene. The encoded poly-glutamine stretch renders mutant huntingtin prone to aggregation. HdhQ150 mice genocopy a pathogenic repeat (∼150 CAGs) in the endogenous mouse huntingtin gene and model predominantly pre-manifest HD. Treating early is likely important to prevent or delay HD, and HdhQ150 mice may be useful to assess therapeutic strategies targeting pre-manifest HD. This requires appropriate markers and here we demonstrate, that pre-symptomatic HdhQ150 mice show several dramatic mutant huntingtin gene-dose dependent pathological changes including: (i) an increase of neuronal intra-nuclear inclusions (NIIs) in brain, (ii) an increase of extra-nuclear aggregates in dentate gyrus, (iii) a decrease of DARPP32 protein and (iv) an increase in glial markers of neuroinflammation, which curiously did not correlate with local neuronal mutant huntingtin inclusion-burden. HdhQ150 mice developed NIIs also in all retinal neuron cell-types, demonstrating that retinal NIIs are not specific to human exon-1 R6 HD mouse models. Taken together, the striking and robust mutant huntingtin gene-dose related changes in aggregate-load, DARPP32 levels and glial activation markers should greatly facilitate future testing of therapeutic strategies in the HdhQ150 HD mouse model.
Collapse
|
132
|
Perucho J, Casarejos MJ, Gómez A, Ruíz C, Fernández-Estevez MÁ, Muñoz MP, de Yébenes JG, Mena MÁ. Striatal infusion of glial conditioned medium diminishes huntingtin pathology in r6/1 mice. PLoS One 2013; 8:e73120. [PMID: 24069174 PMCID: PMC3771920 DOI: 10.1371/journal.pone.0073120] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 07/24/2013] [Indexed: 01/02/2023] Open
Abstract
Huntington's disease is a neurodegenerative disorder caused by an expansion of CAG repeats in the huntingtin gene which produces widespread neuronal and glial pathology. We here investigated the possible therapeutic role of glia or glial products in Huntington's disease using striatal glial conditioned medium (GCM) from fetus mice (E16) continuously infused for 15 and 30 days with osmotic minipumps into the left striatum of R6/1 mice. Animals infused with GCM had significantly less huntingtin inclusions in the ipsilateral cerebral cortex and in the ipsilateral and contralateral striata than mice infused with cerebrospinal fluid. The numbers of DARPP-32 and TH positive neurons were also greater in the ipsilateral but not contralateral striata and substantia nigra, respectively, suggesting a neuroprotective effect of GCM on efferent striatal and nigro-striatal dopamine neurons. GCM increases activity of the autophagic pathway, as shown by the reduction of autophagic substrate, p-62, and the augmentation of LC3 II, Beclin-1 and LAMP-2 protein levels, direct markers of autophagy, in GCM infused mice. GCM also increases BDNF levels. These results suggest that CGM should be further explored as a putative neuroprotective agent in Huntington's disease.
Collapse
Affiliation(s)
- Juan Perucho
- Department of Neurobiology, Hospital “Ramón y Cajal”, Madrid, Spain
- CIBERNED, Madrid, Spain
| | - Maria José Casarejos
- Department of Neurobiology, Hospital “Ramón y Cajal”, Madrid, Spain
- CIBERNED, Madrid, Spain
| | - Ana Gómez
- Department of Neurobiology, Hospital “Ramón y Cajal”, Madrid, Spain
- CIBERNED, Madrid, Spain
| | - Carolina Ruíz
- Department of Neurology, Hospital “Ramón y Cajal”, Madrid, Spain
- CIBERNED, Madrid, Spain
| | | | - Maria Paz Muñoz
- Department of Neurobiology, Hospital “Ramón y Cajal”, Madrid, Spain
- CIBERNED, Madrid, Spain
| | | | - Maria Ángeles Mena
- Department of Neurobiology, Hospital “Ramón y Cajal”, Madrid, Spain
- CIBERNED, Madrid, Spain
- * E-mail:
| |
Collapse
|
133
|
Abstract
Neurodegenerative disorders leading to dementia are common diseases that affect many older and some young adults. Neuroimaging methods are important tools for assessing and monitoring pathological brain changes associated with progressive neurodegenerative conditions. In this review, the authors describe key findings from neuroimaging studies (magnetic resonance imaging and radionucleotide imaging) in neurodegenerative disorders, including Alzheimer's disease (AD) and prodromal stages, familial and atypical AD syndromes, frontotemporal dementia, amyotrophic lateral sclerosis with and without dementia, Parkinson's disease with and without dementia, dementia with Lewy bodies, Huntington's disease, multiple sclerosis, HIV-associated neurocognitive disorder, and prion protein associated diseases (i.e., Creutzfeldt-Jakob disease). The authors focus on neuroimaging findings of in vivo pathology in these disorders, as well as the potential for neuroimaging to provide useful information for differential diagnosis of neurodegenerative disorders.
Collapse
Affiliation(s)
- Shannon L. Risacher
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, and Indiana Alzheimer Disease Center Indiana University School of Medicine, Indianapolis, Indiana
| | - Andrew J. Saykin
- Center for Neuroimaging, Department of Radiology and Imaging Sciences, and Indiana Alzheimer Disease Center Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
134
|
Default-mode network changes in Huntington's disease: an integrated MRI study of functional connectivity and morphometry. PLoS One 2013; 8:e72159. [PMID: 23977239 PMCID: PMC3747049 DOI: 10.1371/journal.pone.0072159] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 07/07/2013] [Indexed: 01/18/2023] Open
Abstract
Previous MRI studies of functional connectivity in pre-symptomatic mutation carriers of Huntington’s disease (HD) have shown dysfunction of the Default-Mode Network (DMN). No data however are currently available on the DMN alterations in the symptomatic stages of the disease, which are characterized by cortical atrophy involving several DMN nodes. We assessed DMN integrity and its possible correlations with motor and cognitive symptoms in 26 symptomatic HD patients as compared to 22 normal volunteers, by analyzing resting state functional MRI data, using the Precuneal Cortex/Posterior Cingulate Cortices (PC/PCC) as seed, controlling at voxel level for the effect of atrophy by co-varying for gray matter volume. Direct correlation with PC/PCC was decreased, without correlation with atrophy, in the ventral medial prefrontal cortex (including anterior cingulate and subgenual cortex), right dorso-medial prefrontal cortex, and in the right inferior parietal cortex (mainly involving the angular gyrus). Negative correlations with PC/PCC were decreased bilaterally in the inferior parietal cortices, while a cluster in the right middle occipital gyrus presented increased correlation with PC/PCC. DMN changes in the ventral medial prefrontal cortex significantly correlated with the performance at the Stroop test (p = .0002). Widespread DMN changes, not correlating with the atrophy of the involved nodes, are present in symptomatic HD patients, and correlate with cognitive disturbances.
Collapse
|
135
|
Trapani A, Palazzo C, de Candia M, Lasorsa FM, Trapani G. Targeting of the Translocator Protein 18 kDa (TSPO): A Valuable Approach for Nuclear and Optical Imaging of Activated Microglia. Bioconjug Chem 2013; 24:1415-28. [DOI: 10.1021/bc300666f] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Adriana Trapani
- Department of Pharmacy and Drug
Sciences, University of Bari, Bari, 70125,
Italy
| | - Claudio Palazzo
- Department of Pharmacy and Drug
Sciences, University of Bari, Bari, 70125,
Italy
| | - Modesto de Candia
- Department of Pharmacy and Drug
Sciences, University of Bari, Bari, 70125,
Italy
| | | | - Giuseppe Trapani
- Department of Pharmacy and Drug
Sciences, University of Bari, Bari, 70125,
Italy
| |
Collapse
|
136
|
Hua J, Unschuld PG, Margolis RL, van Zijl PCM, Ross CA. Elevated arteriolar cerebral blood volume in prodromal Huntington's disease. Mov Disord 2013; 29:396-401. [PMID: 23847161 DOI: 10.1002/mds.25591] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 05/24/2013] [Accepted: 05/29/2013] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Neurovascular alterations have been implicated in the pathophysiology of Huntington's disease (HD). Because arterioles are most responsive to metabolic alterations, arteriolar cerebral blood volume (CBVa) is an important indicator of cerebrovascular regulation. The objective of this pilot study was to investigate potential neurovascular (CBVa ) abnormality in prodromal-HD patients and compare it with the widely used imaging marker: brain atrophy. METHODS CBVa and brain volumes were measured with ultra-high-field (7.0-Telsa) magnetic resonance imaging in seven prodromal-HD patients and nine age-matched controls. RESULTS Cortical CBVa was elevated significantly in prodromal-HD patients compared with controls (relative difference, 38.5%; effect size, 1.48). Significant correlations were found between CBVa in the frontal cortex and genetic measures. By contrast, no significant brain atrophy was detected in the prodromal-HD patients. CONCLUSIONS CBVa may be abnormal in prodromal-HD, even before substantial brain atrophy occurs. Further investigation with a larger cohort and longitudinal follow-up is merited to determine whether CBVa could be used as a potential biomarker for clinical trials.
Collapse
Affiliation(s)
- Jun Hua
- The Russell H. Morgan Department of Radiology and Radiological Science, Division of Magnetic Resonance Research, The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, Maryland, USA
| | | | | | | | | |
Collapse
|
137
|
Lu DY, Huang BR, Yeh WL, Lin HY, Huang SS, Liu YS, Kuo YH. Anti-neuroinflammatory Effect of a Novel Caffeamide Derivative, KS370G, in Microglial cells. Mol Neurobiol 2013; 48:863-74. [DOI: 10.1007/s12035-013-8474-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 05/07/2013] [Indexed: 11/29/2022]
|
138
|
Wu C, Li F, Niu G, Chen X. PET imaging of inflammation biomarkers. Theranostics 2013; 3:448-66. [PMID: 23843893 PMCID: PMC3706689 DOI: 10.7150/thno.6592] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Accepted: 05/24/2013] [Indexed: 01/04/2023] Open
Abstract
Inflammation plays a significant role in many disease processes. Development in molecular imaging in recent years provides new insight into the diagnosis and treatment evaluation of various inflammatory diseases and diseases involving inflammatory process. Positron emission tomography using (18)F-FDG has been successfully applied in clinical oncology and neurology and in the inflammation realm. In addition to glucose metabolism, a variety of targets for inflammation imaging are being discovered and utilized, some of which are considered superior to FDG for imaging inflammation. This review summarizes the potential inflammation imaging targets and corresponding PET tracers, and the applications of PET in major inflammatory diseases and tumor associated inflammation. Also, the current attempt in differentiating inflammation from tumor using PET is also discussed.
Collapse
|
139
|
Scahill R. Recent advances in imaging the onset and progression of Huntington’s disease. Neurodegener Dis Manag 2013. [DOI: 10.2217/nmt.13.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY Huntington’s disease is a devastating autosomal-dominant neurodegenerative disorder resulting in progressive decline in motor and cognitive function, accompanied by neuropsychiatric disturbances. In vivo imaging can reveal the underlying neuropathological changes that contribute to symptom manifestation. Observational studies of individuals carrying the causative gene have demonstrated that structural and functional brain changes are apparent decades before clinical onset of the disease; imaging measures can predict those individuals who subsequently undergo clinical conversion. Such studies have improved our understanding of neurodegeneration across the disease spectrum and aided the identification of therapeutic targets. Clinical trials of potentially disease-modifying treatments are likely to be investigated in the near future and imaging provides a powerful tool to monitor disease progression and thereby assess therapeutic efficacy.
Collapse
Affiliation(s)
- Rachael Scahill
- Huntington’s Disease Research Group, Department of Neurodegeneration, UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
| |
Collapse
|
140
|
Hult Lundh S, Nilsson N, Soylu R, Kirik D, Petersén Å. Hypothalamic expression of mutant huntingtin contributes to the development of depressive-like behavior in the BAC transgenic mouse model of Huntington's disease. Hum Mol Genet 2013; 22:3485-97. [PMID: 23697793 DOI: 10.1093/hmg/ddt203] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Psychiatric symptoms such as depression and anxiety are important clinical features of Huntington's disease (HD). However, the underlying neurobiological substrate for the psychiatric features is not fully understood. In order to explore the biological origin of depression and anxiety in HD, we used a mouse model that expresses the human full-length mutant huntingtin, the BACHD mouse. We found that the BACHD mice displayed depressive- and anxiety-like features as early as at 2 months of age as assessed using the Porsolt forced swim test (FST), the sucrose preference test and the elevated plus maze (EPM). BACHD mice subjected to chronic treatment with the anti-depressant sertraline were not different to vehicle-treated BACHD mice in the FST and EPM. The behavioral manifestations occurred in the absence of reduced hippocampal cell proliferation/neurogenesis or upregulation of the hypothalamic-pituitary-adrenal axis. However, alterations in anxiety- and depression-regulating genes were present in the hypothalamus of BACHD mice including reduced mRNA expression of neuropeptide Y, tachykinin receptor 3 and vesicular monoamine transporter type 2 as well as increased expression of cocaine and amphetamine regulated transcript. Interestingly, the orexin neuronal population in the hypothalamus was increased and showed cellular atrophy in old BACHD mice. Furthermore, inactivation of mutant huntingtin in a subset of the hypothalamic neurons prevented the development of the depressive features. Taken together, our data demonstrate that the BACHD mouse recapitulates clinical HD with early psychiatric aspects and point to the role of hypothalamic dysfunction in the development of depression and anxiety in the disease.
Collapse
Affiliation(s)
- Sofia Hult Lundh
- Translational Neuroendocrine Research Unit, Department of Experimental Medical Science, Lund University, Lund SE-22184, Sweden
| | | | | | | | | |
Collapse
|
141
|
Predominance of D2 receptors in mediating dopamine's effects in brain metabolism: effects of alcoholism. J Neurosci 2013; 33:4527-35. [PMID: 23467368 DOI: 10.1523/jneurosci.5261-12.2013] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Dopamine signals through D1-like and D2-like receptors, which can stimulate or inhibit, respectively, neuronal activity. Here we assessed the balance between D1 or D2 receptor signaling in the human brain and how it is affected in alcoholism. Using PET, we measured the relationship between changes in dopamine and brain glucose metabolism induced by methylphenidate in controls and alcoholics. We show that methylphenidate induced significant DA increases in striatum, amygdala, and medial orbitofrontal cortex, whereas it decreased metabolism in these brain regions. Methylphenidate-induced dopamine increases were greater in controls than in alcoholics, whereas methylphenidate-induced metabolic decreases were greater in alcoholics. For both groups, methylphenidate-induced dopamine increases were associated with decreases in regional brain metabolism, and the correlations were strongest in subthalamic nuclei, anterior cingulate, and medial orbitofrontal cortex. These correlations were more extensive and robust and the slopes steeper in alcoholics than in controls despite their attenuated dopamine responses to methylphenidate, which suggests an impaired modulation of dopamine signals in the brain of alcoholic subjects. These findings are consistent with a predominant inhibitory effect of dopamine in the human brain that is likely mediated by the prominence of dopamine D2/D3 receptors.
Collapse
|
142
|
Younger J, Noor N, McCue R, Mackey S. Low-dose naltrexone for the treatment of fibromyalgia: findings of a small, randomized, double-blind, placebo-controlled, counterbalanced, crossover trial assessing daily pain levels. ACTA ACUST UNITED AC 2013; 65:529-38. [PMID: 23359310 DOI: 10.1002/art.37734] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2012] [Accepted: 09/27/2012] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To determine whether low dosages (4.5 mg/day) of naltrexone reduce fibromyalgia severity as compared with the nonspecific effects of placebo. In this replication and extension study of a previous clinical trial, we tested the impact of low-dose naltrexone on daily self-reported pain. Secondary outcomes included general satisfaction with life, positive mood, sleep quality, and fatigue. METHODS Thirty-one women with fibromyalgia participated in the randomized, double-blind, placebo-controlled, counterbalanced, crossover study. During the active drug phase, participants received 4.5 mg of oral naltrexone daily. An intensive longitudinal design was used to measure daily levels of pain. RESULTS When contrasting the condition end points, we observed a significantly greater reduction of baseline pain in those taking low-dose naltrexone than in those taking placebo (28.8% reduction versus 18.0% reduction; P = 0.016). Low-dose naltrexone was also associated with improved general satisfaction with life (P = 0.045) and with improved mood (P = 0.039), but not improved fatigue or sleep. Thirty-two percent of participants met the criteria for response (defined as a significant reduction in pain plus a significant reduction in either fatigue or sleep problems) during low-dose naltrexone therapy, as contrasted with an 11% response rate during placebo therapy (P = 0.05). Low-dose naltrexone was rated equally tolerable as placebo, and no serious side effects were reported. CONCLUSION The preliminary evidence continues to show that low-dose naltrexone has a specific and clinically beneficial impact on fibromyalgia pain. The medication is widely available, inexpensive, safe, and well-tolerated. Parallel-group randomized controlled trials are needed to fully determine the efficacy of the medication.
Collapse
Affiliation(s)
- Jarred Younger
- Stanford University School of Medicine, Palo Alto, California 94304-1573, USA.
| | | | | | | |
Collapse
|
143
|
Hsiao HY, Chen YC, Chen HM, Tu PH, Chern Y. A critical role of astrocyte-mediated nuclear factor-κB-dependent inflammation in Huntington's disease. Hum Mol Genet 2013; 22:1826-42. [PMID: 23372043 DOI: 10.1093/hmg/ddt036] [Citation(s) in RCA: 162] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Huntington's disease (HD) is an autosomal disease caused by a CAG repeat expansion in the huntingtin (HTT) gene. The resultant mutant HTT protein (mHTT) forms aggregates in various types of cells, including neurons and glial cells and preferentially affects brain function. We found that two HD mouse models (Hdh(150Q) and R6/2) were more susceptible than wild-type (WT) mice to lipopolysaccharide-evoked systemic inflammation and produced more proinflammatory cytokines in the brain. Such an enhanced inflammatory response in the brain was not observed in N171- 82Q mice that express mHTT only in neurons, but not in glial cells. Thus, HD glia might play an important role in chronic inflammation that accelerates disease progression in HD mice. Intriguingly, enhanced activation of nuclear factor (NF)-κB-p65 (p65), a transcriptional mediator of inflammatory responses, was observed in astrocytes of patients and mice with HD. Results obtained using primary R6/2 astrocytes suggest that these cells exhibited higher IκB kinase (IKK) activity that caused prolongation of NF-κB activation, thus upregulating proinflammatory factors during inflammation. R6/2 astrocytes also produced a more-damaging effect on primary R6/2 neurons than did WT astrocytes during inflammation. Blockage of IKK reduced the neuronal toxicity caused by R6/2 astrocytes and ameliorated several HD symptoms of R6/2 mice (e.g. decreased neuronal density, impaired motor coordination and poor cognitive function). Collectively, our results indicate that enhancement of the p65-mediated inflammatory response in astrocytes contributes to HD pathogenesis. Therapeutic interventions aimed at preventing neuronal inflammation may be an important strategy for treating HD.
Collapse
Affiliation(s)
- Han-Yun Hsiao
- Institute of Neuroscience, National Yang-Ming University, Taipei 112, Taiwan
| | | | | | | | | |
Collapse
|
144
|
Georgiou-Karistianis N, Scahill R, Tabrizi SJ, Squitieri F, Aylward E. Structural MRI in Huntington's disease and recommendations for its potential use in clinical trials. Neurosci Biobehav Rev 2013; 37:480-90. [PMID: 23376047 DOI: 10.1016/j.neubiorev.2013.01.022] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 01/02/2013] [Accepted: 01/22/2013] [Indexed: 01/18/2023]
Abstract
Huntington's disease (HD) results in progressive impairment of motor and cognitive function and neuropsychiatric disturbance. There are no disease-modifying treatments available, but HD research is entering a critical phase where promising disease-specific therapies are on the horizon. Thus, a pressing need exists for biomarkers capable of monitoring progression and ultimately determining drug efficacy. Neuroimaging provides a powerful tool for assessing disease progression. However, in order to be accepted as biomarkers for clinical trials, imaging measures must be reproducible, robust to scanner differences, sensitive to disease-related change and demonstrate a relationship to clinically meaningful measures. We provide a review of the current structural imaging literature in HD and highlight inconsistencies between studies. We make recommendations for the standardisation of reporting for future studies, such as appropriate cohort characterisation and documentation of methodologies to facilitate comparisons and inform trial design. We also argue for an intensified effort to consider issues highlighted here so that we have the best chance of assessing the efficacy of the therapeutic benefit in forestalling this devastating disease.
Collapse
|
145
|
Positron emission tomography imaging in neurological disorders. J Neurol 2013; 259:1769-80. [PMID: 22297461 DOI: 10.1007/s00415-012-6428-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 01/16/2012] [Accepted: 01/18/2012] [Indexed: 01/28/2023]
Abstract
Positron emission tomography (PET) is a powerful tool for in vivo imaging investigations of human brain function. It provides non-invasive quantification of brain metabolism, receptor binding of various neurotransmitter systems, and alterations in regional blood flow. The use of PET in a clinical setting is still limited due to the high costs of cyclotrons and radiochemical laboratories. However, once these limitations can be bypassed, PET could aid clinical practice by providing a useful imaging technique for the diagnosis, the planning of treatment, and the prediction outcome in various neurological diseases.This review aims to explain the PET imaging technique and its applications in neurological disorders such as Parkinson’s disease, Huntington’s disease, multiple sclerosis, and dementias.
Collapse
|
146
|
Kwan W, Träger U, Davalos D, Chou A, Bouchard J, Andre R, Miller A, Weiss A, Giorgini F, Cheah C, Möller T, Stella N, Akassoglou K, Tabrizi SJ, Muchowski PJ. Mutant huntingtin impairs immune cell migration in Huntington disease. J Clin Invest 2012; 122:4737-47. [PMID: 23160193 DOI: 10.1172/jci64484] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 09/27/2012] [Indexed: 11/17/2022] Open
Abstract
In Huntington disease (HD), immune cells are activated before symptoms arise; however, it is unclear how the expression of mutant huntingtin (htt) compromises the normal functions of immune cells. Here we report that primary microglia from early postnatal HD mice were profoundly impaired in their migration to chemotactic stimuli, and expression of a mutant htt fragment in microglial cell lines was sufficient to reproduce these deficits. Microglia expressing mutant htt had a retarded response to a laser-induced brain injury in vivo. Leukocyte recruitment was defective upon induction of peritonitis in HD mice at early disease stages and was normalized upon genetic deletion of mutant htt in immune cells. Migration was also strongly impaired in peripheral immune cells from pre-manifest human HD patients. Defective actin remodeling in immune cells expressing mutant htt likely contributed to their migration deficit. Our results suggest that these functional changes may contribute to immune dysfunction and neurodegeneration in HD, and may have implications for other polyglutamine expansion diseases in which mutant proteins are ubiquitously expressed.
Collapse
Affiliation(s)
- Wanda Kwan
- Biomedical Sciences Program, UCSF, San Francisco, California, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
147
|
Figiel M, Szlachcic WJ, Switonski PM, Gabka A, Krzyzosiak WJ. Mouse models of polyglutamine diseases: review and data table. Part I. Mol Neurobiol 2012; 46:393-429. [PMID: 22956270 PMCID: PMC3461215 DOI: 10.1007/s12035-012-8315-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2012] [Accepted: 07/29/2012] [Indexed: 12/23/2022]
Abstract
Polyglutamine (polyQ) disorders share many similarities, such as a common mutation type in unrelated human causative genes, neurological character, and certain aspects of pathogenesis, including morphological and physiological neuronal alterations. The similarities in pathogenesis have been confirmed by findings that some experimental in vivo therapy approaches are effective in multiple models of polyQ disorders. Additionally, mouse models of polyQ diseases are often highly similar between diseases with respect to behavior and the features of the disease. The common features shared by polyQ mouse models may facilitate the investigation of polyQ disorders and may help researchers explore the mechanisms of these diseases in a broader context. To provide this context and to promote the understanding of polyQ disorders, we have collected and analyzed research data about the characterization and treatment of mouse models of polyQ diseases and organized them into two complementary Excel data tables. The data table that is presented in this review (Part I) covers the behavioral, molecular, cellular, and anatomic characteristics of polyQ mice and contains the most current knowledge about polyQ mouse models. The structure of this data table is designed in such a way that it can be filtered to allow for the immediate retrieval of the data corresponding to a single mouse model or to compare the shared and unique aspects of many polyQ models. The second data table, which is presented in another publication (Part II), covers therapeutic research in mouse models by summarizing all of the therapeutic strategies employed in the treatment of polyQ disorders, phenotypes that are used to examine the effects of the therapy, and therapeutic outcomes.
Collapse
Affiliation(s)
- Maciej Figiel
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland.
| | | | | | | | | |
Collapse
|
148
|
Abstract
Here, we synthesize research behind the emerging hypothesis that inflammation--which can result, for example, from viral infections--can initiate and propagate chronic neuronal dysfunction, an event that precedes the clinical onset of many neurodegenerative diseases. Therapeutic approaches that target immunological pathways in the prodromal phase of diseases might decrease the incidence of neurodegenerative disorders and increase the therapeutic window for neuroprotection.
Collapse
|
149
|
Ruiz C, Casarejos MJ, Gomez A, Solano R, de Yebenes JG, Mena MA. Protection by glia-conditioned medium in a cell model of Huntington disease. PLOS CURRENTS 2012; 4:e4fbca54a2028b. [PMID: 22919565 PMCID: PMC3423315 DOI: 10.1371/4fbca54a2028b] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The physiological role of huntingtin and the pathogenic mechanisms that produce the disease are unknown. Mutant huntingtin changes its normal localization and produces cytoplasmic and intranuclear inclusions, changes gene transcription, alters synaptic transmission, impairs mitochondrial activity and activates caspases and other pro-apoptotic molecules, promotes excitotoxicity, energy deficits, synthesis and release reduction of neurotrophic factors and oxidative stress. Previous studies confirm that the mutant huntingtin difficult neurotrophic function of astrocytes leading to neuronal dysfunction in Huntington’s disease. Our objective was to study the neuroprotective potential role of glia-conditioned medium (GCM) in an in vitro model of Huntington’s disease. We used conditionally-immortalized striatal neuronal progenitor cell lines (STHdhQ7/Q7 and STHdhQ111/Q111) expressing endogenous levels of normal and mutant huntingtin with 7 and 111 glutamines, respectively. We studied the protection of fetal and postnatal glia conditioned medium (GCM) on H2O2 (2 µM), glutamate (5 mM) and 3-nitropropionic acid (2.5 mM) related toxicity. We also compared the neuroprotective effects of GCM versus that of the growth factors bFGF, BDNF and GDNF.
Fetal GCM protects from every toxin, reducing the cell death and increasing the cell survival. Fetal GCM reduces the caspases fragmentation of the protein PARP, the expression of chaperone Hsp70 and the accumulation of ROS and polyubiquitinated proteins. In addition, in Q111 striatal cells treated with H2O2 (2 µM) for 24 hours, the intracellular GSH levels are higher in the presence of GCM. Notably, the 13-day and 2-month postnatal GCM, totally protects from H2O2 induced cell death in mutant striatal cells. GCM neuroprotective effects are more potent than those of the already identified neurotrophic factors.
We conclude that GCM protects Q111 cells from neuronal neurotoxins and the effects of GCM are more potent than those of any known neurotrophic factor. GCM may contain new and more potent, as yet unidentified, neurotrophic molecules, potentially useful in patients with Huntington’s disease.
Collapse
|
150
|
Etemad S, Zamin RM, Ruitenberg MJ, Filgueira L. A novel in vitro human microglia model: characterization of human monocyte-derived microglia. J Neurosci Methods 2012; 209:79-89. [PMID: 22659341 DOI: 10.1016/j.jneumeth.2012.05.025] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Revised: 04/20/2012] [Accepted: 05/22/2012] [Indexed: 12/18/2022]
Abstract
Microglia are the innate immune cells of the central nervous system. They help maintaining physiological homeostasis and contribute significantly to inflammatory responses in the course of infection, injury and degenerative processes. To date, there is no standardized simple model available to investigate the biology of human microglia. The aim of this study was to establish a new human microglia model. For that purpose, human peripheral blood monocytes were cultured in serum free medium in the presence of M-CSF, GM-CSF, NGF and CCL2 to generate monocyte-derived microglia (M-MG). M-MG were clearly different in morphology, phenotype and function from freshly isolated monocytes, cultured monocytes in the absence of the cytokines and monocyte-derived dendritic cells (M-DC) cultured in the presence of GM-CSF and IL-4. M-MG acquired a ramified morphology with primary and secondary processes. M-MG displayed a comparable phenotype to the human microglia cell line HMC3, expressing very low levels of CD45, CD14 and HLA-DR, CD11b and CD11c; and undetectable levels of CD40, CD80 and CD83, and a distinct pattern of chemokine receptors (positive for CCR1, CCR2, CCR4, CCR5, CXCR1, CXCR3, CX3CR1; negative for CCR6 and CCR7). In comparison with M-DC, M-MG displayed lower T-lymphocyte stimulatory capacity, as well as lower phagocytosis activity. The described protocol for the generation of human monocyte-derived microglia is feasible, well standardized and reliable, as it uses well defined culture medium and recombinant cytokines, but no serum or conditioned medium. This protocol will certainly be very helpful for future studies investigating the biology and pathology of human microglia.
Collapse
Affiliation(s)
- Samar Etemad
- School of Anatomy, Physiology and Human Biology, University of Western Australia, WA 6009, Australia
| | | | | | | |
Collapse
|