51
|
Jones CD, Motl R, Sandroff BM. Depression in multiple sclerosis: Is one approach for its management enough? Mult Scler Relat Disord 2021; 51:102904. [PMID: 33780807 DOI: 10.1016/j.msard.2021.102904] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/18/2021] [Accepted: 03/15/2021] [Indexed: 01/04/2023]
Abstract
BACKGROUND Major depression disorder (MDD) and severe depression symptoms are highly prevalent in multiple sclerosis (MS). Depression can worsen symptoms of MS and is associated with significantly reduced quality of life and increased risk of suicide. Currently, there is no gold-standard, single treatment available for depression in MS. Pharmacotherapy, cognitive behavior therapy (CBT), and exercise training individually are moderately, yet incompletely, efficacious for managing depression in the general population and MS. PURPOSE This review provides an overview of evidence from meta-analyses and systematic reviews for current treatments of depression in persons with MS. This review further develops the rationale for using a combinatory treatment approach in persons with MS. METHODS We performed a narrative review of meta-analyses and systematic reviews regarding the current state of evidence for the three most common treatments of depression in persons with MS (i.e., antidepressant medication, cognitive-behavior therapy, and exercise training). We provide a concise assessment of the overall effect of these treatments on depression in the general population and then persons with MS. We further note short-comings of research on these treatments for depression. CONCLUSION There is no single, gold-standard treatment for depression in MS, and we proposed that combinatory treatments should be considered for the management of depression in MS. However, there is a paucity of evidence for the use of combinatory therapy on depression and its outcomes in persons with MS, and this supports direct examination of the feasibility and efficacy of such combinatory approaches for MDD in MS.
Collapse
Affiliation(s)
- C Danielle Jones
- University of Alabama at Birmingham, Department of Physical Therapy, Birmingham, AL, United States.
| | - Robert Motl
- University of Alabama at Birmingham, Department of Physical Therapy, Birmingham, AL, United States
| | - Brian M Sandroff
- Kessler Foundation, Center for Neuropsychology and Neuroscience, West Orange, NJ, United States
| |
Collapse
|
52
|
Zhang L, Hu K, Shao T, Hou L, Zhang S, Ye W, Josephson L, Meyer JH, Zhang MR, Vasdev N, Wang J, Xu H, Wang L, Liang SH. Recent developments on PET radiotracers for TSPO and their applications in neuroimaging. Acta Pharm Sin B 2021; 11:373-393. [PMID: 33643818 PMCID: PMC7893127 DOI: 10.1016/j.apsb.2020.08.006] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/15/2020] [Accepted: 07/29/2020] [Indexed: 12/12/2022] Open
Abstract
The 18 kDa translocator protein (TSPO), previously known as the peripheral benzodiazepine receptor, is predominately localized to the outer mitochondrial membrane in steroidogenic cells. Brain TSPO expression is relatively low under physiological conditions, but is upregulated in response to glial cell activation. As the primary index of neuroinflammation, TSPO is implicated in the pathogenesis and progression of numerous neuropsychiatric disorders and neurodegenerative diseases, including Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), multiple sclerosis (MS), major depressive disorder (MDD) and obsessive compulsive disorder (OCD). In this context, numerous TSPO-targeted positron emission tomography (PET) tracers have been developed. Among them, several radioligands have advanced to clinical research studies. In this review, we will overview the recent development of TSPO PET tracers, focusing on the radioligand design, radioisotope labeling, pharmacokinetics, and PET imaging evaluation. Additionally, we will consider current limitations, as well as translational potential for future application of TSPO radiopharmaceuticals. This review aims to not only present the challenges in current TSPO PET imaging, but to also provide a new perspective on TSPO targeted PET tracer discovery efforts. Addressing these challenges will facilitate the translation of TSPO in clinical studies of neuroinflammation associated with central nervous system diseases.
Collapse
Key Words
- AD, Alzheimer's disease
- ALS, amyotrophic lateral sclerosis
- AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid
- ANT, adenine nucleotide transporter
- Am, molar activities
- BBB, blood‒brain barrier
- BMSC, bone marrow stromal cells
- BP, binding potential
- BPND, non-displaceable binding potential
- BcTSPO, Bacillus cereus TSPO
- CBD, corticobasal degeneration
- CNS disorders
- CNS, central nervous system
- CRAC, cholesterol recognition amino acid consensus sequence
- DLB, Lewy body dementias
- EP, epilepsy
- FTD, frontotemporal dementia
- HAB, high-affinity binding
- HD, Huntington's disease
- HSE, herpes simplex encephalitis
- IMM, inner mitochondrial membrane
- KA, kainic acid
- LAB, low-affinity binding
- LPS, lipopolysaccharide
- MAB, mixed-affinity binding
- MAO-B, monoamine oxidase B
- MCI, mild cognitive impairment
- MDD, major depressive disorder
- MMSE, mini-mental state examination
- MRI, magnetic resonance imaging
- MS, multiple sclerosis
- MSA, multiple system atrophy
- Microglial activation
- NAA/Cr, N-acetylaspartate/creatine
- Neuroinflammation
- OCD, obsessive compulsive disorder
- OMM, outer mitochondrial membrane
- P2X7R, purinergic receptor P2X7
- PAP7, RIa-associated protein
- PBR, peripheral benzodiazepine receptor
- PCA, posterior cortical atrophy
- PD, Parkinson's disease
- PDD, PD dementia
- PET, positron emission tomography
- PKA, protein kinase A
- PRAX-1, PBR-associated protein 1
- PSP, progressive supranuclear palsy
- Positron emission tomography (PET)
- PpIX, protoporphyrin IX
- QA, quinolinic acid
- RCYs, radiochemical yields
- ROS, reactive oxygen species
- RRMS, relapsing remitting multiple sclerosis
- SA, specific activity
- SAH, subarachnoid hemorrhage
- SAR, structure–activity relationship
- SCIDY, spirocyclic iodonium ylide
- SNL, selective neuronal loss
- SNR, signal to noise ratio
- SUV, standard uptake volume
- SUVR, standard uptake volume ratio
- TBAH, tetrabutyl ammonium hydroxide
- TBI, traumatic brain injury
- TLE, temporal lobe epilepsy
- TSPO
- TSPO, translocator protein
- VDAC, voltage-dependent anion channel
- VT, distribution volume
- d.c. RCYs, decay-corrected radiochemical yields
- dMCAO, distal middle cerebral artery occlusion
- fP, plasma free fraction
- n.d.c. RCYs, non-decay-corrected radiochemical yields
- p.i., post-injection
Collapse
Affiliation(s)
- Lingling Zhang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
- Department of Neurology, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Kuan Hu
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Tuo Shao
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Lu Hou
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Shaojuan Zhang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Weijian Ye
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Lee Josephson
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Jeffrey H. Meyer
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health & Department of Psychiatry, University of Toronto, Toronto ON M5T 1R8, Canada
| | - Ming-Rong Zhang
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Neil Vasdev
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
- Azrieli Centre for Neuro-Radiochemistry, Brain Health Imaging Centre, Centre for Addiction and Mental Health & Department of Psychiatry, University of Toronto, Toronto ON M5T 1R8, Canada
| | - Jinghao Wang
- Department of Pharmacy, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Hao Xu
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, Guangzhou 510630, China
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Steven H. Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
53
|
Raimo S, Santangelo G, Trojano L. The emotional disorders associated with multiple sclerosis. HANDBOOK OF CLINICAL NEUROLOGY 2021; 183:197-220. [PMID: 34389118 DOI: 10.1016/b978-0-12-822290-4.00009-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Multiple sclerosis (MS) is associated with a high prevalence of emotional and mood disorders. Emotional disorders may worsen during illness progression and affect the quality of life of patients and their families. MS is often associated with depression, with an increased risk of suicide, poor adherence to treatment, decreased functional status, and quality of life. The diagnosis and treatment of emotional and mood disorders in these patients is often challenging since several symptoms of these disorders overlap with those of MS. Other prevalent emotional disorders in MS include bipolar disorder, anxiety disorders, emotional blunting (apathy), and pseudobulbar affect. Early recognition and treatment of these comorbidities could contribute to the reduction of disability and even to decreased mortality. The aim of this chapter is to provide an up-to-date review of mood and emotional disorders that are often associated with MS, focusing on their epidemiology, clinical features, pathogenesis, assessment, and treatment. The interplay between the psychosocial impact of the chronic disability and the demyelinating structural lesions of the brain in precipitating emotional and mood disorders is discussed, as well as its implications for diagnosis and treatment.
Collapse
Affiliation(s)
- Simona Raimo
- Department of Psychology, University of Campania "Luigi Vanvitelli", Caserta, Italy.
| | - Gabriella Santangelo
- Department of Psychology, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Luigi Trojano
- Department of Psychology, University of Campania "Luigi Vanvitelli", Caserta, Italy
| |
Collapse
|
54
|
Lapato AS, Thompson SM, Parra K, Tiwari-Woodruff SK. Astrocyte Glutamate Uptake and Water Homeostasis Are Dysregulated in the Hippocampus of Multiple Sclerosis Patients With Seizures. ASN Neuro 2020; 12:1759091420979604. [PMID: 33297722 PMCID: PMC7734542 DOI: 10.1177/1759091420979604] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
While seizure disorders are more prevalent among multiple sclerosis (MS) patients than the population overall and prognosticate earlier death & disability, their etiology remains unclear. Translational data indicate perturbed expression of astrocytic molecules contributing to homeostatic neuronal excitability, including water channels (AQP4) and synaptic glutamate transporters (EAAT2), in a mouse model of MS with seizures (MS+S). However, astrocytes in MS+S have not been examined. To assess the translational relevance of astrocyte dysfunction observed in a mouse model of MS+S, demyelinated lesion burden, astrogliosis, and astrocytic biomarkers (AQP4/EAAT2/ connexin-CX43) were evaluated by immunohistochemistry in postmortem hippocampi from MS & MS+S donors. Lesion burden was comparable in MS & MS+S cohorts, but astrogliosis was elevated in MS+S CA1 with a concomitant decrease in EAAT2 signal intensity. AQP4 signal declined in MS+S CA1 & CA3 with a loss of perivascular AQP4 in CA1. CX43 expression was increased in CA3. Together, these data suggest that hippocampal astrocytes from MS+S patients display regional differences in expression of molecules associated with glutamate buffering and water homeostasis that could exacerbate neuronal hyperexcitability. Importantly, mislocalization of CA1 perivascular AQP4 seen in MS+S is analogous to epileptic hippocampi without a history of MS, suggesting convergent pathophysiology. Furthermore, as neuropathology was concentrated in MS+S CA1, future study is warranted to determine the pathophysiology driving regional differences in glial function in the context of seizures during demyelinating disease.
Collapse
Affiliation(s)
- Andrew S Lapato
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, California, United States.,Center for Glial-Neuronal Interaction, UCR School of Medicine, Riverside, California, United States
| | - Sarah M Thompson
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, California, United States
| | - Karen Parra
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, California, United States
| | - Seema K Tiwari-Woodruff
- Division of Biomedical Sciences, UCR School of Medicine, Riverside, California, United States.,Center for Glial-Neuronal Interaction, UCR School of Medicine, Riverside, California, United States.,Department of Neuroscience, UCR School of Medicine, Riverside, California, United States
| |
Collapse
|
55
|
Heitmann H, Andlauer TFM, Korn T, Mühlau M, Henningsen P, Hemmer B, Ploner M. Fatigue, depression, and pain in multiple sclerosis: How neuroinflammation translates into dysfunctional reward processing and anhedonic symptoms. Mult Scler 2020; 28:1020-1027. [PMID: 33179588 PMCID: PMC9131410 DOI: 10.1177/1352458520972279] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Fatigue, depression, and pain affect the majority of multiple sclerosis
(MS) patients, which causes a substantial burden to patients and
society. The pathophysiology of these symptoms is not entirely clear,
and current treatments are only partially effective. Clinically, these
symptoms share signs of anhedonia, such as reduced motivation and a
lack of positive affect. In the brain, they are associated with
overlapping structural and functional alterations in areas involved in
reward processing. Moreover, neuroinflammation has been shown to
directly impede monoaminergic neurotransmission that plays a key role
in reward processing. Here, we review recent neuroimaging and
neuroimmunological findings, which indicate that dysfunctional reward
processing might represent a shared functional mechanism fostering the
symptom cluster of fatigue, depression, and pain in MS. We propose a
framework that integrates these findings with a focus on monoaminergic
neurotransmission and discuss its therapeutic implications,
limitations, and perspectives.
Collapse
Affiliation(s)
- Henrik Heitmann
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany/TUM-Neuroimaging Center, School of Medicine, Technical University of Munich, Munich, Germany/Department of Psychosomatic Medicine and Psychotherapy, School of Medicine, Technical University of Munich, Munich, Germany
| | - Till F M Andlauer
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Thomas Korn
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany/ Department of Experimental Neuroimmunology, Technical University of Munich, Munich, Germany/Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Mark Mühlau
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany/TUM-Neuroimaging Center, School of Medicine, Technical University of Munich, Munich, Germany
| | - Peter Henningsen
- Department of Psychosomatic Medicine and Psychotherapy, School of Medicine, Technical University of Munich, Munich, Germany
| | - Bernhard Hemmer
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany/Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Markus Ploner
- Department of Neurology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany/TUM-Neuroimaging Center, School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
56
|
Chen T, Liu S, Zheng M, Li Y, He L. The effect of geniposide on chronic unpredictable mild stress‐induced depressive mice through
BTK
/
TLR4
/
NF‐κB
and
BDNF
/
TrkB
signaling pathways. Phytother Res 2020; 35:932-945. [DOI: 10.1002/ptr.6846] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/15/2020] [Accepted: 07/18/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Tong Chen
- Department of Pharmacology China Pharmaceutical University Nanjing China
| | - Shengnan Liu
- Department of Pharmacology China Pharmaceutical University Nanjing China
| | - Menglin Zheng
- Department of Pharmacology China Pharmaceutical University Nanjing China
| | - Yixuan Li
- Department of Pharmacology China Pharmaceutical University Nanjing China
| | - Ling He
- Department of Pharmacology China Pharmaceutical University Nanjing China
| |
Collapse
|
57
|
Martino M, Magioncalda P, El Mendili MM, Droby A, Paduri S, Schiavi S, Petracca M, Inglese M. Depression is associated with disconnection of neurotransmitter-related nuclei in multiple sclerosis. Mult Scler 2020; 27:1102-1111. [PMID: 32907463 DOI: 10.1177/1352458520948214] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Depression is frequently associated with multiple sclerosis (MS). However, the biological background underlying such association is poorly understood. OBJECTIVE Investigating the functional connections of neurotransmitter-related brainstem nuclei, along with their relationship with white matter (WM) microstructure, in MS patients with depressive symptomatology (MS-D) and without depressive symptomatology (MS-nD). METHODS Combined resting-state functional magnetic resonance imaging (fMRI) and diffusion-weighted MRI (dMRI) study on 50 MS patients, including 19 MS-D and 31 MS-nD patients, along with 37 healthy controls (HC). Main analyses performed are (1) comparison between groups of raphe nuclei (RN)-related functional connectivity (FC); (2) correlation between RN-related FC and whole brain dMRI-derived fractional anisotropy (FA) map; and (3) comparison between groups of FA in the RN-related WM area. RESULTS (1) RN-related FC was reduced in MS-D when compared to MS-nD and HC; (2) RN-related FC positively correlated with FA in a WM cluster mainly encompassing thalamic/basal ganglia regions, including the fornix; and (3) FA in such WM area was reduced in MS-D. CONCLUSION Depressive symptomatology in MS is specifically associated to a functional disconnection of neurotransmitter-related nuclei, which in turn may be traced to a distinct spatial pattern of WM alterations mainly involving the limbic network.
Collapse
Affiliation(s)
- Matteo Martino
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paola Magioncalda
- Graduate Institute of Mind, Brain, and Consciousness, Taipei Medical University, Taipei, Taiwan/Brain and Consciousness Research Center, Taipei Medical University - Shuang Ho Hospital, New Taipei City, Taiwan/Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Psychiatry, University of Genoa, Genoa, Italy
| | | | - Amgad Droby
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Swetha Paduri
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Simona Schiavi
- Department of Computer Science, University of Verona, Verona, Italy/Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health Section of Neurology, University of Genoa, Genoa, Italy
| | - Maria Petracca
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matilde Inglese
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA/Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Section of Neurology, University of Genoa, Genoa, Italy/Ospedale Policlinico San Martino IRCCS, Genoa, Italy
| |
Collapse
|
58
|
Wei F, Wu M, Qian S, Li D, Jin M, Wang J, Shui L, Lin H, Tang M, Chen K. Association between short-term exposure to ambient air pollution and hospital visits for depression in China. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 724:138207. [PMID: 32268289 DOI: 10.1016/j.scitotenv.2020.138207] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/06/2020] [Accepted: 03/23/2020] [Indexed: 06/11/2023]
Abstract
Depression is one of the leading causes of disability, but the etiology remains unclear. Recently, it has been suggested that air pollution is a potential risk factor for depression. However, the results remained inconsistent. So we conducted this study to assess the association between short-term exposure to ambient air pollution and hospital visits for depression in China. Daily hospital visits for depression from January 18, 2013 to June 10, 2018 were extracted from a regional health information system (HIS) covered 1.34 million population in Ningbo, China. We collected daily air pollutant concentrations and meteorological data from environmental air quality monitoring sites and meteorological stations in the study area. Quasi-Poisson regression models with generalized additive models (GAM) were applied to explore the associations between air pollution and hospital visits for depression. Stratified analyses were also conducted by gender, age, and season to examine the effects modification. The results disclosed that air pollutants including PM2.5, PM10, SO2, CO, and NO2 were positively correlated with hospital visits for depression. The strongest effects all occurred on lag0 (the same) day, and the corresponding excess risks (ERs) were 2.59 (95%CI: 0.72, 4.49) for PM2.5, 3.08 (95%CI: 1.05, 5.16) for PM10, 3.22 (95%CI: 1.16, 5.32) for SO2, 4.38 (95%CI: 1.83, 6.99) for CO, and 4.94 (95%CI: 2.03, 7.92) for NO2 per IQR increase, respectively. The associations were found to be stronger in the elderly (≥65 years) and cold season. Furthermore, the effects of CO and NO2 remained significant in most two-pollutant models, suggesting that traffic-related air pollutants might be more important triggers of depression.
Collapse
Affiliation(s)
- Fang Wei
- Department of Epidemiology and Biostatistics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Mengyin Wu
- Department of Epidemiology and Biostatistics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Sangni Qian
- Department of Epidemiology and Biostatistics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Die Li
- Department of Epidemiology and Biostatistics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Mingjuan Jin
- Department of Epidemiology and Biostatistics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Jianbing Wang
- Department of Epidemiology and Biostatistics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Liming Shui
- Health Commission of Ningbo, Zhejiang, China
| | - Hongbo Lin
- The Center for Disease Control and Prevention of Yinzhou District, Ningbo, Zhejiang, China
| | - Mengling Tang
- Department of Epidemiology and Biostatistics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Kun Chen
- Department of Epidemiology and Biostatistics, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China; Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China.
| |
Collapse
|
59
|
Glukhovsky L, Brandstadter R, Leavitt VM, Krieger S, Buyukturkoglu K, Fabian M, Sand IK, Klineova S, Riley CS, Lublin FD, Miller AE, Sumowski JF. Hippocampal volume is more related to patient-reported memory than objective memory performance in early multiple sclerosis. Mult Scler 2020; 27:568-578. [PMID: 32567468 DOI: 10.1177/1352458520922830] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND When persons with multiple sclerosis (MS) report memory decline but objective memory performance is normal, there is a bias toward believing objective test results. OBJECTIVE Investigate whether subjective memory decline or objective memory performance is more related to hippocampal and hippocampal subfield volumes in early MS. METHODS Persons with early MS (n = 185; ⩽5.0 years diagnosed) completed a subjective memory questionnaire; an objective memory composite was derived from four memory tests. Total hippocampal and subfield volumes were derived from high-resolution 3.0 T magnetic resonance images (MRIs). Partial correlations assessed links between hippocampal volumes and both subjective and objective memory, controlling for age, sex, mood, and pre-morbid intelligence quotient (IQ). RESULTS Lower total hippocampal and CA1 volumes were related to worse subjective memory but not objective memory (controlling for multiple comparisons). Correlations between subjective memory and both CA1 and subiculum were significantly stronger than were correlations between objective memory and these subfields. Patients in the worst tertile of subjective memory complaints (but not objective memory) had lower hippocampal volumes than 35 demographically similar healthy controls. CONCLUSION Patient-report is inherently a longitudinal assessment of within-person memory change in everyday life, which may be more sensitive to subtle disease-related changes than cross-sectional objective tests. Findings align with the aging literature.
Collapse
Affiliation(s)
- Lisa Glukhovsky
- The Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Victoria M Leavitt
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Stephen Krieger
- The Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Korhan Buyukturkoglu
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Michelle Fabian
- The Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ilana Katz Sand
- The Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sylvia Klineova
- The Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Claire S Riley
- Department of Neurology, Columbia University Irving Medical Center, New York, NY, USA
| | - Fred D Lublin
- The Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aaron E Miller
- The Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - James F Sumowski
- The Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
60
|
Ouallet JC, Radat F, Creange A, Abdellaoui M, Heinzlef O, Giannesini C, Hautecoeur P, Lamargue Hamel D, Deloire M, Brochet B, Jean Deleglise AS, Lehert P. Evaluation of emotional disorders before and during treatment with interferon beta in patients with multiple sclerosis. J Neurol Sci 2020; 413:116739. [PMID: 32151852 DOI: 10.1016/j.jns.2020.116739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 02/14/2020] [Accepted: 02/15/2020] [Indexed: 10/25/2022]
Abstract
BACKGROUND Domains encompassing emotional disorders in relapsing-remitting MS (RRMS) patients are still unclear. METHODS We performed a 24-month, multicenter, single-arm, prospective study. RRMS patients started IFN-β treatment at baseline. The primary endpoint was lack of emotional control, measured using the "Echelle d'HumeurDépressive" (EHD) scale three times at baseline and at 10 post-treatment visits. Secondary endpoints were emotional blunting, irritability, fatigue, depression and anxiety. A linear mixed covariance model assessed change from baseline on an intention-to-treat basis, under the assumption of no mood disorder effect (one-sided 97.5% level), in which autoregressive type of autocorrelation was tested. RESULTS Out of 79 recruited patients, 70 were analyzed: 80% female; mean (SD) age, 37.0 (11.5) years. Mean (SD) lack of emotional control score at baseline and Month 24 was 12.7 (4.4) and 12.6 (5.5), respectively, versus 10.1 (3.2) in a healthy control population matched for age and sex. Stepwise analysis identified younger age, male sex and antidepressant use as significant predictors of higher lack of emotional control values. CONCLUSIONS Based on 24 months of prospective follow-up, the results of this study highlights a broad spectrum of emotional disorders in the MS population at the time of disease modifying drugs initiation but no major IFN-β-related emotional disorders (mood dyscontrol, anxiety, depression) were observed. However, sporadic occurrences of severe mood disorders and suicidality cannot be excluded.
Collapse
Affiliation(s)
- Jean-Christophe Ouallet
- Service de Neurologie, Centre Hospitalier Universitaire (CHU) de Bordeaux, Bordeaux, France.
| | - Françoise Radat
- Service de Neurologie, Centre Hospitalier Universitaire (CHU) de Bordeaux, Bordeaux, France
| | - Alain Creange
- Service de Neurologie, Groupe Hospitalier Henri Mondor, APHP, Université Paris Est-Créteil, Creteil, France
| | - Mohamed Abdellaoui
- Service de Neurologie, Groupe Hospitalier Henri Mondor, APHP, Université Paris Est-Créteil, Creteil, France
| | - Olivier Heinzlef
- Service de Neurologie, Centre Hospitalier de Poissy, Poissy, France
| | - Claire Giannesini
- Service de Neurologie, Centre Hospitalier Universitaire St Antoine, APHP, Paris, France
| | | | | | - Mathilde Deloire
- Service de Neurologie, Centre Hospitalier Universitaire (CHU) de Bordeaux, Bordeaux, France
| | - Bruno Brochet
- Service de Neurologie, Centre Hospitalier Universitaire (CHU) de Bordeaux, Bordeaux, France; NeuroCentre Magendie, INSERM U862, University of Bordeaux, Bordeaux, France
| | | | - Philippe Lehert
- Faculty of Economics, Louvain University, Belgium; Faculty of Medicine, University of Melbourne, Australia
| |
Collapse
|
61
|
Czysz A. Impact of Medical Comorbidity in Biomarker Discovery for Major Depressive Disorder. Psychiatr Ann 2020. [DOI: 10.3928/00485713-20200507-02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
62
|
Jadrešin O, Sila S, Trivić I, Mišak Z, Kolaček S, Hojsak I. Lactobacillus reuteri DSM 17938 is effective in the treatment of functional abdominal pain in children: Results of the double-blind randomized study. Clin Nutr 2020; 39:3645-3651. [PMID: 32362486 DOI: 10.1016/j.clnu.2020.04.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/05/2020] [Accepted: 04/08/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Possible therapeutic effect of Lactobacillus (L.) reuteri DSM 17938 has been reported in children with functional abdominal pain (FAP) but data are inconclusive. METHODS This is a randomized double-blinded controlled trial (RCT) which assessed effect of L. reuteri DSM 17938 (dose 108 CFU/day) in children (age 4-18 years) on FAP during an intervention period of 12 weeks and follow-up of 4 weeks. This study was performed after the interim analysis and had different labeling of products and a new randomization. Data presented here are results of this RCT and pooled data from both RCTs (before and after interim analysis). RESULTS This RCT included 46 children (median age 10.1 vs 10.6 years; 11 vs 13 girls). Abdominal pain was less severe in intervention group during the 4th month of the study and there was significant increase in the number of days without pain. Pooled data from both parts of the study included 101 children. Number of days without pain was significantly higher in the L. reuteri group (mean difference 26.42 days, 95% CI 22.47-30.17). Significant difference in the pain intensity was found after 2nd, 3rd and 4th month of the intervention. There was no difference between groups in the number of children in whom symptoms completely ceased (Risk Ratio 1.09, 95% CI 0.75-1.58). CONCLUSION Administration of L. reuteri DSM 17938 was associated with the reduction in the intensity of pain and with significantly increase in pain-free days in children with FAP.
Collapse
Affiliation(s)
- Oleg Jadrešin
- Referral Center for Pediatric Gastroenterology and Nutrition, Children's Hospital Zagreb, Zagreb, Croatia
| | - Sara Sila
- Referral Center for Pediatric Gastroenterology and Nutrition, Children's Hospital Zagreb, Zagreb, Croatia
| | - Ivana Trivić
- Referral Center for Pediatric Gastroenterology and Nutrition, Children's Hospital Zagreb, Zagreb, Croatia
| | - Zrinjka Mišak
- Referral Center for Pediatric Gastroenterology and Nutrition, Children's Hospital Zagreb, Zagreb, Croatia
| | - Sanja Kolaček
- Referral Center for Pediatric Gastroenterology and Nutrition, Children's Hospital Zagreb, Zagreb, Croatia; University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Iva Hojsak
- Referral Center for Pediatric Gastroenterology and Nutrition, Children's Hospital Zagreb, Zagreb, Croatia; University of Zagreb, School of Medicine, Zagreb, Croatia; University J.J. Strossmayer Osijek, School of Medicine, Osijek, Croatia.
| |
Collapse
|
63
|
Wang Y, Tian F, Fitzgerald KC, Bhattarai JJ, Naismith RT, Hyland M, Calabresi PA, Mowry EM. Socioeconomic status and race are correlated with affective symptoms in multiple sclerosis. Mult Scler Relat Disord 2020; 41:102010. [PMID: 32088654 DOI: 10.1016/j.msard.2020.102010] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 02/13/2020] [Indexed: 12/01/2022]
Abstract
OBJECTIVE Investigate the relationship between socioeconomic status (SES) and race with self-reported fatigue, depression, and anxiety levels in multiple sclerosis (MS). METHODS Cross-sectional review of the MS Partners Advancing Technology and Health Solutions (MS PATHS) database for adults with MS in the United States. We evaluated race and socioeconomic status (available markers: insurance, employment status, or level of education) as predictors of fatigue, depression, and anxiety sub-scores of the Neuro-QoL (Quality of life in neurological disorders), with particular interest between Caucasians/whites (CA) and African Americans/blacks (AA). Multivariate linear regression models included as covariates age, sex, disability status, smoking status, body mass index, and disease-modifying therapy. RESULTS 7,430 individuals were included; compared to CA, AA tended to be younger, more female-predominant, and had a higher level of disability. AA had completed slightly less education, had a higher level of Medicaid coverage or uninsured status, and had higher rates of unemployed or disabled status. In the univariate model, markers of lower SES, by whichever definition we used, correlated with worse affective symptoms. In the multivariate model stratified by race, CA showed similar trends. In contrast, in AA, only lower SES by employment status was correlated with worse affective symptoms. In both CA and AA, moderate and severe level of disability correlated with worse affective symptoms. CONCLUSION SES and race may influence affective symptoms reported by individuals with MS. The reasons for the correlation are likely multifactorial. Longitudinal studies should strive to identify factors associated with risk of affective symptoms in MS that may be modifiable.
Collapse
Affiliation(s)
- Yujie Wang
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N. Wolfe St.Pathology 627, Baltimore, MD 21287, USA.
| | - Fan Tian
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N. Wolfe St.Pathology 627, Baltimore, MD 21287, USA
| | - Kathryn C Fitzgerald
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N. Wolfe St.Pathology 627, Baltimore, MD 21287, USA
| | - Jagriti Jackie Bhattarai
- Department of Physical Medicine and Rehabilitation, Johns Hopkins University School of Medicine, USA
| | - Robert T Naismith
- Department of Neurology, Washington University in St. Louis School of Medicine, USA
| | - Megan Hyland
- Department of Neurology, University of Rochester School of Medicine, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N. Wolfe St.Pathology 627, Baltimore, MD 21287, USA
| | - Ellen M Mowry
- Department of Neurology, Johns Hopkins University School of Medicine, 600 N. Wolfe St.Pathology 627, Baltimore, MD 21287, USA
| |
Collapse
|
64
|
das Neves SP, Santos G, Barros C, Pereira DR, Ferreira R, Mota C, Monteiro S, Fernandes A, Marques F, Cerqueira JJ. Enhanced cognitive performance in experimental autoimmune encephalomyelitis mice treated with dimethyl fumarate after the appearance of disease symptoms. J Neuroimmunol 2020; 340:577163. [PMID: 31982706 DOI: 10.1016/j.jneuroim.2020.577163] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 01/02/2020] [Accepted: 01/10/2020] [Indexed: 12/23/2022]
Abstract
In multiple sclerosis (MS), cognitive dysfunction is common but difficult to treat. We analyzed the impact of dimethyl fumarate, an MS drug with neuroprotective properties, in spatial memory performance in a mouse model of MS and looked for structural correlates in the hippocampus. Treated mice presented better cognitive performance which was not associated with structural hippocampal damage but with decreased demyelination in the fimbria. Dimethyl fumarate, even if initiated after hindlimb paralysis, ameliorated memory deficits in the MS mouse model due, at least in part, to its positive impact in the demyelination of the main hippocampal output pathway.
Collapse
Affiliation(s)
- Sofia P das Neves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Gisela Santos
- Neuron-Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Catarina Barros
- Neuron-Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Diana Rodrigues Pereira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Ricardo Ferreira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Cristina Mota
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Susana Monteiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Adelaide Fernandes
- Neuron-Glia Biology in Health and Disease, Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Portugal; Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Portugal
| | - Fernanda Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - João José Cerqueira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; 2CA, Clinical Academic Center - Braga, Braga, Portugal.
| |
Collapse
|
65
|
Microglial activation in schizophrenia: Is translocator 18 kDa protein (TSPO) the right marker? Schizophr Res 2020; 215:167-172. [PMID: 31699629 DOI: 10.1016/j.schres.2019.10.045] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/12/2022]
Abstract
Positron emission tomography (PET) with translocator 18 kDa protein (TSPO) radioligands has frequently been used to investigate microglial activation in schizophrenia in vivo. However, the specificity of this marker is increasingly debated. Here we show that TSPO expression is 1) not increased in postmortem brain tissue of schizophrenia patients; 2) not correlated with expression of microglial activation markers; 3) not restricted to microglia; and 4) not upregulated in ex vivo activated human primary microglia. Our data are in line with recent reports showing that TSPO expression is not increased in schizophrenia and that it is not a specific marker for activated microglia. This study emphasizes the need for further development of tracers to study the role of microglial activation in schizophrenia and other diseases.
Collapse
|
66
|
Gut microbiota depletion from early adolescence alters adult immunological and neurobehavioral responses in a mouse model of multiple sclerosis. Neuropharmacology 2019; 157:107685. [DOI: 10.1016/j.neuropharm.2019.107685] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/21/2019] [Accepted: 06/24/2019] [Indexed: 02/06/2023]
|
67
|
Singhal T, O'Connor K, Dubey S, Pan H, Chu R, Hurwitz S, Cicero S, Tauhid S, Silbersweig D, Stern E, Kijewski M, DiCarli M, Weiner HL, Bakshi R. Gray matter microglial activation in relapsing vs progressive MS: A [F-18]PBR06-PET study. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2019; 6:e587. [PMID: 31355321 PMCID: PMC6624145 DOI: 10.1212/nxi.0000000000000587] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 05/15/2019] [Indexed: 11/15/2022]
Abstract
Objective To determine the value of [F-18]PBR06-PET for assessment of microglial activation in the cerebral gray matter in patients with MS. Methods Twelve patients with MS (7 relapsing-remitting and 5 secondary progressive [SP]) and 5 healthy controls (HCs) had standardized uptake value (SUV) PET maps coregistered to 3T MRI and segmented into cortical and subcortical gray matter regions. SUV ratios (SUVRs) were global brain normalized. Voxel-by-voxel analysis was performed using statistical parametric mapping (SPM). Normalized brain parenchymal volumes (BPVs) were determined from MRI using SIENAX. Results Cortical SUVRs were higher in the hippocampus, amygdala, midcingulate, posterior cingulate, and rolandic operculum and lower in the medial-superior frontal gyrus and cuneus in the MS vs HC group (all p < 0.05). Subcortical gray matter SUVR was higher in SPMS vs RRMS (+10.8%, p = 0.002) and HC (+11.3%, p = 0.055) groups. In the MS group, subcortical gray matter SUVR correlated with the Expanded Disability Status Scale (EDSS) score (r = 0.75, p = 0.005) and timed 25-foot walk (T25FW) (r = 0.70, p = 0.01). Thalamic SUVRs increased with increasing EDSS scores (r = 0.83, p = 0.0008) and T25FW (r = 0.65, p = 0.02) and with decreasing BPV (r = -0.63, p = 0.03). Putaminal SUVRs increased with increasing EDSS scores (0.71, p = 0.009) and with decreasing BPV (r = -0.67, p = 0.01). On SPM analysis, peak correlations of thalamic voxels with BPV were seen in the pulvinar and with the EDSS score and T25FW in the dorsomedial thalamic nuclei. Conclusions This study suggests that [F-18]PBR06-PET detects widespread abnormal microglial activation in the cerebral gray matter in MS. Increased translocator protein binding in subcortical gray matter regions is associated with brain atrophy and may link to progressive MS.
Collapse
Affiliation(s)
- Tarun Singhal
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Kelsey O'Connor
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Shipra Dubey
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Hong Pan
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Renxin Chu
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Shelley Hurwitz
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Steven Cicero
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Shahamat Tauhid
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - David Silbersweig
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Emily Stern
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Marie Kijewski
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Marcelo DiCarli
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Howard L Weiner
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Rohit Bakshi
- Partners MS Center (T.S., K.O.C., R.C., S.C., S.T., H.L.W., R.B.), Laboratory for Neuroimaging Research, Ann Romney Center for Neurological Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School; Division of Nuclear Medicine and Molecular Imaging (S.D., M.K., M.D.), Department of Radiology, Brigham and Women's Hospital, Harvard Medical School; Functional Neuroimaging Laboratory (H.P., D.S., E.S.), Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School; Department of Medicine (S.H.) and Department of Radiology (E.S., R.B.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
68
|
Abstract
Cognitive impairment is increasingly recognized to be a core feature of multiple sclerosis (MS), with important implications for the everyday life of individuals with MS and for disease management. Unfortunately, the exact mechanisms that underlie this cognitive impairment are poorly understood and there are no effective therapeutic options for this aspect of the disease. During MS, focal brain inflammatory lesions, together with pathological changes of both CNS grey matter and normal-appearing white matter, can interfere with cognitive functions. Moreover, inflammation may alter the crosstalk between the immune and the nervous systems, modulating the induction of synaptic plasticity and neurotransmission. In this Review, we examine the CNS structures and cognitive domains that are affected by the disease, with a specific focus on hippocampal involvement in MS and experimental autoimmune encephalomyelitis, an experimental model of MS. We also discuss the hypothesis that, during MS, immune-mediated alterations of synapses' ability to express long-term plastic changes may contribute to the pathogenesis of cognitive impairment by interfering with the dynamics of neuronal networks.
Collapse
|
69
|
Duarte-Silva E, Macedo D, Maes M, Peixoto CA. Novel insights into the mechanisms underlying depression-associated experimental autoimmune encephalomyelitis. Prog Neuropsychopharmacol Biol Psychiatry 2019; 93:1-10. [PMID: 30849414 DOI: 10.1016/j.pnpbp.2019.03.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 02/28/2019] [Accepted: 03/03/2019] [Indexed: 02/07/2023]
Abstract
Multiple Sclerosis (MS) is a chronic autoimmune disease characterized by neuroinflammation, demyelination and neuroaxonal degeneration affecting >2 million people around the world. MS is often accompanied by psychiatric comorbidities such as major depressive disorder (MDD), which presents a lifetime prevalence of around 50% in MS patients. Experimental Autoimmune Encephalomyelitis (EAE) is an animal model extensively used to study MS. EAE mimics the autoimmune nature of MS, as well as its inflammatory and demyelinating mechanisms also presenting predictive validity. There are important similarities between EAE and MS-associated depression (MSD). The mechanisms shared by these disorders include peripheral inflammation, neuroinflammation, mitochondrial dysfunctions, oxidative stress, nitrosative stress, lowered antioxidant defenses, increased bacterial translocation into the systemic circulation, and microglial pathology. Although the role of the immune-inflammatory system in MDD has been established in the 1990's, only few studies addressed immune pathways as a major determinant of depressive-like behavior in EAE. Therefore, in the present study we aimed at revising the current literature on EAE as an animal model to investigate the comorbidity between MS and MDD. In this regard, we revised the current literature on behavioral alterations in EAE, the possible mechanisms involved in this comorbidity and the potential and limitations of using this animal model to study depressive-like behavior.
Collapse
Affiliation(s)
- Eduardo Duarte-Silva
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Recife, PE, Brazil; Postgraduate Program in Biosciences and Biotechnology for Health (PPGBBS), Oswaldo Cruz Foundation (FIOCRUZ-PE)/Aggeu Magalhães Institute (IAM), Recife, PE, Brazil.
| | - Danielle Macedo
- Neuropsychopharmacology Laboratory, Drug Research and Development Center, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; Department of Physiology and Pharmacology, Faculty of Medicine, Universidade Federal do Ceará, Fortaleza, CE, Brazil; National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, Brazil
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Christina Alves Peixoto
- Laboratory of Ultrastructure, Aggeu Magalhães Institute (IAM), Recife, PE, Brazil; Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil.
| |
Collapse
|
70
|
Yang J, Liu R, Lu F, Xu F, Zheng J, Li Z, Cui W, Wang C, Zhang J, Xu S, Zhou W, Wang Q, Chen J, Chen X. Fast Green FCF Attenuates Lipopolysaccharide-Induced Depressive-Like Behavior and Downregulates TLR4/Myd88/NF-κB Signal Pathway in the Mouse Hippocampus. Front Pharmacol 2019; 10:501. [PMID: 31139084 PMCID: PMC6519320 DOI: 10.3389/fphar.2019.00501] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 04/23/2019] [Indexed: 01/09/2023] Open
Abstract
Depression is a common neuropsychiatric disorder and new anti-depressive treatments are still in urgent demand. Fast Green FCF, a safe biocompatible color additive, has been suggested to mitigate chronic pain. However, Fast green FCF’s effect on depression is unknown. We aimed to investigate Fast green FCF’s effect on lipopolysaccharide (LPS)-induced depressive-like behavior and the underlying mechanisms. Pretreatment of Fast green FCF (100 mg/kg, i.p. daily for 7 days) alleviated depressive-like behavior in LPS-treated mice. Fast green FCF suppressed the LPS-induced microglial and astrocyte activation in the hippocampus. Fast green FCF decreased the mRNA and protein levels of Toll-like receptor 4 (TLR4) and Myeloid differentiation primary response 88 (Myd88) and suppressed the phosphorylation of nuclear factor-κB (NF-κB) in the hippocampus of LPS-treated mice. Fast green FCF also downregulated hippocampal tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6, but did not alter the level of the brain-derived neurotrophic factor (BDNF) in the hippocampus of LPS-treated mice. The molecular docking simulation predicts that Fast green FCF may interact with TLR4 and interrupt the formation of the TLR4-MD2 complex. In conclusion, the anti-depressive action of Fast green FCF in LPS-treated mice may involve the suppression of neuroinflammation and the downregulation of TLR4/Myd88/NF-κB signal pathway in mouse hippocampus. Our findings indicate the potential of Fast green FCF for controlling depressive symptoms.
Collapse
Affiliation(s)
- Jing Yang
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Rongjun Liu
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Fan Lu
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Fang Xu
- Department of Anesthesiology, Ningbo No. 2 Hospital, Ningbo, China
| | - Jinwei Zheng
- Department of Anesthesiology, Ningbo No. 2 Hospital, Ningbo, China
| | - Zhao Li
- Department of Anesthesiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wei Cui
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Chuang Wang
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Junfang Zhang
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Shujun Xu
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Wenhua Zhou
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Qinwen Wang
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| | - Junping Chen
- Department of Anesthesiology, Ningbo No. 2 Hospital, Ningbo, China
| | - Xiaowei Chen
- Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, Ningbo, China
| |
Collapse
|
71
|
Corallo F, Lo Buono V, Genovese R, Palmeri R, Di Cara M, Rifici C, Sessa E, D'Aleo G, Galletti F, Bonanno L, Marino S. A complex relation between depression and multiple sclerosis: a descriptive review. Neurol Sci 2019; 40:1551-1558. [PMID: 31001715 DOI: 10.1007/s10072-019-03889-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Accepted: 04/08/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) is a demyelinating neurodegenerative disease that affects central nervous system (CNS). MS patients are more likely to develop depressive symptoms than patients with other chronic diseases. OBJECTIVE In this review, we have analysed if there is a correlation between brain lesions (BL), structural damage (SD) and depressive symptoms (DS). METHODS We Searched on PubMed and Web of Science databases and screening references of included studied and some review article for additional citations. From initial 745 studies, only 9 met the inclusion criteria. All studies conducted research on 389 patients with MS associated with DS and 120 HC (healthy controls). RESULTS The selected researches highlighted the involvement of limbic system, the role of hippocampus and the impact of brain lesions on the emotional status of MS patients. DISCUSSION In the genesis of depression are implicated many mechanisms including genetic, biochemical, immunological and psychosocial factors, even if a prominent role in the onset of DS seem to be associated with structural and functional brain alterations.
Collapse
Affiliation(s)
- Francesco Corallo
- IRCCS Centro Neurolesi "Bonino-Pulejo", S.S. 113 Via Palermo, C.da Casazza, 98124, Messina, Italy
| | - Viviana Lo Buono
- IRCCS Centro Neurolesi "Bonino-Pulejo", S.S. 113 Via Palermo, C.da Casazza, 98124, Messina, Italy
| | - Roberto Genovese
- IRCCS Centro Neurolesi "Bonino-Pulejo", S.S. 113 Via Palermo, C.da Casazza, 98124, Messina, Italy
| | - Rosanna Palmeri
- IRCCS Centro Neurolesi "Bonino-Pulejo", S.S. 113 Via Palermo, C.da Casazza, 98124, Messina, Italy
| | - Marcella Di Cara
- IRCCS Centro Neurolesi "Bonino-Pulejo", S.S. 113 Via Palermo, C.da Casazza, 98124, Messina, Italy
| | - Carmela Rifici
- IRCCS Centro Neurolesi "Bonino-Pulejo", S.S. 113 Via Palermo, C.da Casazza, 98124, Messina, Italy
| | - Edoardo Sessa
- IRCCS Centro Neurolesi "Bonino-Pulejo", S.S. 113 Via Palermo, C.da Casazza, 98124, Messina, Italy
| | - Giangaetano D'Aleo
- IRCCS Centro Neurolesi "Bonino-Pulejo", S.S. 113 Via Palermo, C.da Casazza, 98124, Messina, Italy
| | | | - Lilla Bonanno
- IRCCS Centro Neurolesi "Bonino-Pulejo", S.S. 113 Via Palermo, C.da Casazza, 98124, Messina, Italy
| | - Silvia Marino
- IRCCS Centro Neurolesi "Bonino-Pulejo", S.S. 113 Via Palermo, C.da Casazza, 98124, Messina, Italy.
| |
Collapse
|
72
|
Bauckneht M, Capitanio S, Raffa S, Roccatagliata L, Pardini M, Lapucci C, Marini C, Sambuceti G, Inglese M, Gallo P, Cecchin D, Nobili F, Morbelli S. Molecular imaging of multiple sclerosis: from the clinical demand to novel radiotracers. EJNMMI Radiopharm Chem 2019; 4:6. [PMID: 31659498 PMCID: PMC6453990 DOI: 10.1186/s41181-019-0058-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 03/21/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Brain PET imaging with different tracers is mainly clinically used in the field of neurodegenerative diseases and brain tumors. In recent years, the potential usefulness of PET has also gained attention in the field of MS. In fact, MS is a complex disease and several processes can be selected as a target for PET imaging. The use of PET with several different tracers has been mainly evaluated in the research setting to investigate disease pathophysiology (i.e. phenotypes, monitoring of progression) or to explore its use a surrogate end-point in clinical trials. RESULTS We have reviewed PET imaging studies in MS in humans and animal models. Tracers have been grouped according to their pathophysiological targets (ie. tracers for myelin kinetic, neuroinflammation, and neurodegeneration). The emerging clinical indication for brain PET imaging in the differential diagnosis of suspected tumefactive demyelinated plaques as well as the clinical potential provided by PET images in view of the recent introduction of PET/MR technology are also addressed. CONCLUSION While several preclinical and fewer clinical studies have shown results, full-scale clinical development programs are needed to translate molecular imaging technologies into a clinical reality that could ideally fit into current precision medicine perspectives.
Collapse
Affiliation(s)
- Matteo Bauckneht
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genoa, Italy
| | - Selene Capitanio
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genoa, Italy
| | - Stefano Raffa
- Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
| | - Luca Roccatagliata
- Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
- Neuroradiology, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Matteo Pardini
- Clinical Neurology, Department of Neuroscience (DINOGMI), University of Genoa, Genoa, Italy
- Clinica Neurologica, IRCCS Ospedale Policlinico, San Martino, Genoa, Italy
| | - Caterina Lapucci
- Clinical Neurology, Department of Neuroscience (DINOGMI), University of Genoa, Genoa, Italy
| | - Cecilia Marini
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genoa, Italy
- CNR Institute of Molecular Bioimaging and Physiology, Milan, Italy
| | - Gianmario Sambuceti
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
| | - Matilde Inglese
- Clinical Neurology, Department of Neuroscience (DINOGMI), University of Genoa, Genoa, Italy
- Clinica Neurologica, IRCCS Ospedale Policlinico, San Martino, Genoa, Italy
| | - Paolo Gallo
- Multiple Sclerosis Centre of the Veneto Region, Department of Neurosciences DNS, University of Padua, Padua, Italy
| | - Diego Cecchin
- Nuclear Medicine Unit, Department of Medicine-DIMED, Padova University Hospital, Padua, Italy
- Padua Neuroscience Center, University of Padua, Padua, Italy
| | - Flavio Nobili
- Clinical Neurology, Department of Neuroscience (DINOGMI), University of Genoa, Genoa, Italy
- Clinica Neurologica, IRCCS Ospedale Policlinico, San Martino, Genoa, Italy
| | - Silvia Morbelli
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, 16132 Genoa, Italy
- Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
| |
Collapse
|
73
|
Morris G, Maes M, Berk M, Puri BK. Myalgic encephalomyelitis or chronic fatigue syndrome: how could the illness develop? Metab Brain Dis 2019; 34:385-415. [PMID: 30758706 PMCID: PMC6428797 DOI: 10.1007/s11011-019-0388-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 01/23/2019] [Indexed: 12/19/2022]
Abstract
A model of the development and progression of chronic fatigue syndrome (myalgic encephalomyelitis), the aetiology of which is currently unknown, is put forward, starting with a consideration of the post-infection role of damage-associated molecular patterns and the development of chronic inflammatory, oxidative and nitrosative stress in genetically predisposed individuals. The consequences are detailed, including the role of increased intestinal permeability and the translocation of commensal antigens into the circulation, and the development of dysautonomia, neuroinflammation, and neurocognitive and neuroimaging abnormalities. Increasing levels of such stress and the switch to immune and metabolic downregulation are detailed next in relation to the advent of hypernitrosylation, impaired mitochondrial performance, immune suppression, cellular hibernation, endotoxin tolerance and sirtuin 1 activation. The role of chronic stress and the development of endotoxin tolerance via indoleamine 2,3-dioxygenase upregulation and the characteristics of neutrophils, monocytes, macrophages and T cells, including regulatory T cells, in endotoxin tolerance are detailed next. Finally, it is shown how the immune and metabolic abnormalities of chronic fatigue syndrome can be explained by endotoxin tolerance, thus completing the model.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Victoria, Australia
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Victoria, Australia
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Barwon Health, Deakin University, Geelong, Victoria, Australia
- Department of Psychiatry, Royal Melbourne Hospital, University of Melbourne, Parkville, Victoria, Australia
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
- Orygen, The National Centre of Excellence in Youth Mental Health, Parkville, Victoria, Australia
| | - Basant K Puri
- Department of Medicine, Imperial College London, Hammersmith Hospital, London, England, W12 0HS, UK.
| |
Collapse
|
74
|
Feng M, Crowley NA, Patel A, Guo Y, Bugni SE, Luscher B. Reversal of a Treatment-Resistant, Depression-Related Brain State with the Kv7 Channel Opener Retigabine. Neuroscience 2019; 406:109-125. [PMID: 30858110 DOI: 10.1016/j.neuroscience.2019.03.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 02/28/2019] [Accepted: 03/01/2019] [Indexed: 12/27/2022]
Abstract
Neuroinflammation is associated with increased vulnerability to diverse psychiatric conditions, including treatment-resistant major depressive disorder (MDD). Here we assessed whether high fat diet (HFD) induced neuroinflammation may be suitable to model a treatment-resistant depressive-like brain state in mice. Male and female mice were fed a HFD for 18 weeks, followed by quantitation of glucose tolerance, inflammatory markers of brain tissue (TNFα, IL-6, IL-1β, Iba-1), neural excitability in the prelimbic cortex (PLC), as well as assessment of emotional reactivity and hedonic behavior in a battery of behavioral tests. In addition, we assessed the behavioral responsiveness of mice to fluoxetine, desipramine, ketamine, and the Kv7 channel opener and anticonvulsant retigabine. HFD exposure led to glucose intolerance and neuroinflammation in male mice, with similar but non-significant trends in females. Neuroinflammation of males was associated with anxious-depressive-like behavior and defects in working memory, along with neural hyperexcitability and increased Ih currents of pyramidal cells in the PLC. The behavioral changes were largely resistant to chronic treatment with fluoxetine and desipramine, as well as ketamine. By contrast, retigabine (also known as ezogabine) normalized neural excitability and Ih currents recorded from slices of HFD-treated animals and significantly ameliorated most of the behavioral impairments, without effects in control diet exposed animals. Thus, treatment resistant depressive-like brain states that are associated with chronic neuroinflammation may involve hyperexcitability of pyramidal neurons and may be effectively treated by retigabine.
Collapse
Affiliation(s)
- Mengyang Feng
- Department of Biology, Pennsylvania State University, University Park, PA 16802; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802
| | - Nicole A Crowley
- Department of Biology, Pennsylvania State University, University Park, PA 16802; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802
| | - Akshilkumar Patel
- Department of Biology, Pennsylvania State University, University Park, PA 16802; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802
| | - Yao Guo
- Department of Biology, Pennsylvania State University, University Park, PA 16802; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802
| | - Sierra E Bugni
- Department of Biology, Pennsylvania State University, University Park, PA 16802; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802
| | - Bernhard Luscher
- Department of Biology, Pennsylvania State University, University Park, PA 16802; Department of Biochemistry & Molecular Biology, Pennsylvania State University, University Park, PA 16802; Center for Molecular Investigation of Neurological Disorders (CMIND), The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA 16802.
| |
Collapse
|
75
|
Alò R, Zizza M, Fazzari G, Facciolo RM, Canonaco M. Genistein Modifies Hamster Behavior and Expression of Inflammatory Factors following Subchronic Unpredictable Mild Stress. Neuroendocrinology 2019; 108:98-108. [PMID: 30408789 DOI: 10.1159/000495209] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/08/2018] [Indexed: 11/19/2022]
Abstract
BACKGROUND Previous studies have pointed to the protective role of genistein against stress adaptations although neuromolecular mechanisms are not yet fully known. With this work, we evaluated the influence of such a phytoestrogen on hamster behavioral and molecular activities following exposure to subchronic unpredictable mild stress. METHODS The motor behaviors of hamsters (n = 28) were analyzed using elevated plus maze (EPM) test, hole board (HB) test, and forced swim test (FST). In addition, neurodegeneration events were assessed with amino cupric silver stain, while expression variations of tropomyosin receptor kinase B (TrkB), nuclear factor kappa-B1 (NF-κB1), and heat shock protein 70 (Hsp70) mRNAs were highlighted in limbic neuronal fields via in situ hybridization. RESULTS Genistein accounted for increased motor performances in EPM and HB tests but reduced immobility during FST, which were correlated with diminished argyrophilic signals in some limbic neuronal fields. Contextually, upregulated Hsp70 and TrkB mRNAs occurred in hippocampal (HIP) and hypothalamic neuronal fields. Conversely, diminished NF-κB1 levels were mainly obtained in HIP. CONCLUSION Hormonal neuroprotective properties of genistein corroborating anxiolytic and antidepressant role(s) through elevated expression levels of stress proteins and trophic factors may constitute novel therapeutic measures against emotional and stress-related motor performances.
Collapse
Affiliation(s)
- Raffaella Alò
- Comparative Neuroanatomy Laboratory, Biology, Ecology and Earth Science Department, University of Calabria, Arcavacata di Rende, Italy,
| | - Merylin Zizza
- Comparative Neuroanatomy Laboratory, Biology, Ecology and Earth Science Department, University of Calabria, Arcavacata di Rende, Italy
| | - Gilda Fazzari
- Comparative Neuroanatomy Laboratory, Biology, Ecology and Earth Science Department, University of Calabria, Arcavacata di Rende, Italy
| | - Rosa Maria Facciolo
- Comparative Neuroanatomy Laboratory, Biology, Ecology and Earth Science Department, University of Calabria, Arcavacata di Rende, Italy
| | - Marcello Canonaco
- Comparative Neuroanatomy Laboratory, Biology, Ecology and Earth Science Department, University of Calabria, Arcavacata di Rende, Italy
| |
Collapse
|
76
|
Effect of overnight smoking abstinence on a marker for microglial activation: a [ 11C]DAA1106 positron emission tomography study. Psychopharmacology (Berl) 2018; 235:3525-3534. [PMID: 30343364 PMCID: PMC6497451 DOI: 10.1007/s00213-018-5077-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 10/10/2018] [Indexed: 12/27/2022]
Abstract
RATIONALE Microglia are the main immune cells in the central nervous system and participate in neuroinflammation. When activated, microglia express increased levels of the translocator protein 18 kDa (TSPO), thereby making TSPO availability a marker for neuroinflammation. Using positron emission tomography (PET) scanning, our group recently demonstrated that smokers in the satiated state had 16.8% less binding of the radiotracer [11C]DAA1106 (a radioligand for TSPO) in the brain than nonsmokers. OBJECTIVES We sought to determine the effect of overnight smoking abstinence on [11C]DAA1106 binding in the brain. METHODS Forty participants (22 smokers and 18 nonsmokers) completed the study (at one of two sites) and had usable data, which included images from a dynamic [11C]DAA1106 PET scanning session (with smokers having been abstinent for 17.9 ± 2.3 h) and a blood sample for TSPO genotyping. Whole brain standardized uptake values (SUVs) were determined, and analysis of variance was performed, with group (overnight abstinent smoker vs. nonsmoker), site, and TSPO genotype as factors, thereby controlling for site and genotype. RESULTS Overnight abstinent smokers had lower whole brain SUVs (by 15.5 and 17.0% for the two study sites) than nonsmokers (ANCOVA, P = 0.004). The groups did not significantly differ in injected radiotracer dose or body weight, which were used to calculate SUV. CONCLUSIONS These results in overnight abstinent smokers are similar to those in satiated smokers, indicating that chronic cigarette smoking leads to global impairment of microglial activation which persists into early abstinence. Other explanations for study results, such as smoking leading to reduced numbers of microglia or smokers having more rapid metabolism of the radiotracer than nonsmokers, are also possible.
Collapse
|
77
|
Gao F, Yin X, Edden RA, Evans AC, Xu J, Cao G, Li H, Li M, Zhao B, Wang J, Wang G. Altered hippocampal GABA and glutamate levels and uncoupling from functional connectivity in multiple sclerosis. Hippocampus 2018; 28:813-823. [PMID: 30069963 PMCID: PMC6251738 DOI: 10.1002/hipo.23001] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 05/01/2018] [Accepted: 06/11/2018] [Indexed: 12/16/2022]
Abstract
There is growing evidence for dysfunctional glutamatergic excitation and/or gamma-aminobutyric acid (GABA)ergic inhibition in patients with multiple sclerosis (MS). Cognitive impairment may occur during the early stages of MS and hippocampal abnormalities have been suggested as biomarkers. However, researchers have not clearly determined whether changes in hippocampal GABA and glutamate (Glu) levels are associated with cognitive impairment and aberrant neural activity in patients with MS. We used magnetic resonance spectroscopy to measure GABA+ and Glu levels in the left hippocampal region of 29 patients with relapsing-remitting MS and 29 healthy controls (HCs). Resting-state functional connectivity (FC) with the hippocampus was also examined. Compared to HCs, patients exhibited significantly lower GABA+ and Glu levels, which were associated with verbal and visuospatial memory deficits, respectively. Patients also showed decreased FC strengths between the hippocampus and several cortical regions, which are located within the default mode network. Moreover, hippocampal GABA+ levels and Glu/GABA+ ratios correlated with the FC strengths in HCs but not in patients with MS. This study describes a novel method for investigating the complex relationships among excitatory/inhibitory neurotransmitters, brain connectivity and cognition in health and disease. Strategies that modulate Glu and GABA neurotransmission may represent new therapeutic treatments for patients with MS.
Collapse
Affiliation(s)
- Fei Gao
- Shandong Medical Imaging Research Institute, Shandong University, Jinan, China
| | - Xuntao Yin
- Department of Radiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Richard A.E. Edden
- Russell H. Morgan Department of Radiology and Radiological Science, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
- FM Kirby Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD, USA
| | - Alan C. Evans
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Junhai Xu
- Tianjin Key Laboratory of Cognitive Computing and Application, School of Computer Science and Technology, Tianjin University, Tianjin, China
| | - Guanmei Cao
- Shandong Medical Imaging Research Institute, Shandong University, Jinan, China
| | - Honghao Li
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan 250021, China
| | - Muwei Li
- Vanderbilt University Institute of Imaging Science, Nashville, TN, USA
| | - Bin Zhao
- Shandong Medical Imaging Research Institute, Shandong University, Jinan, China
| | - Jian Wang
- Department of Radiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Guangbin Wang
- Shandong Medical Imaging Research Institute, Shandong University, Jinan, China
| |
Collapse
|
78
|
Comninos AN, Demetriou L, Wall MB, Shah AJ, Clarke SA, Narayanaswamy S, Nesbitt A, Izzi-Engbeaya C, Prague JK, Abbara A, Ratnasabapathy R, Yang L, Salem V, Nijher GM, Jayasena CN, Tanner M, Bassett P, Mehta A, McGonigle J, Rabiner EA, Bloom SR, Dhillo WS. Modulations of human resting brain connectivity by kisspeptin enhance sexual and emotional functions. JCI Insight 2018; 3:121958. [PMID: 30333302 PMCID: PMC6237465 DOI: 10.1172/jci.insight.121958] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 08/17/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Resting brain connectivity is a crucial component of human behavior demonstrated by disruptions in psychosexual and emotional disorders. Kisspeptin, a recently identified critical reproductive hormone, can alter activity in certain brain structures but its effects on resting brain connectivity and networks in humans remain elusive. METHODS We determined the effects of kisspeptin on resting brain connectivity (using functional neuroimaging) and behavior (using psychometric analyses) in healthy men, in a randomized double-blinded 2-way placebo-controlled study. RESULTS Kisspeptin's modulation of the default mode network (DMN) correlated with increased limbic activity in response to sexual stimuli (globus pallidus r = 0.500, P = 0.005; cingulate r = 0.475, P = 0.009). Furthermore, kisspeptin's DMN modulation was greater in men with less reward drive (r = -0.489, P = 0.008) and predicted reduced sexual aversion (r = -0.499, P = 0.006), providing key functional significance. Kisspeptin also enhanced key mood connections including between the amygdala-cingulate, hippocampus-cingulate, and hippocampus-globus pallidus (all P < 0.05). Consistent with this, kisspeptin's enhancement of hippocampus-globus pallidus connectivity predicted increased responses to negative stimuli in limbic structures (including the thalamus and cingulate [all P < 0.01]). CONCLUSION Taken together, our data demonstrate a previously unknown role for kisspeptin in the modulation of functional brain connectivity and networks, integrating these with reproductive hormones and behaviors. Our findings that kisspeptin modulates resting brain connectivity to enhance sexual and emotional processing and decrease sexual aversion, provide foundation for kisspeptin-based therapies for associated disorders of body and mind. FUNDING NIHR, MRC, and Wellcome Trust.
Collapse
Affiliation(s)
- Alexander N Comninos
- Investigative Medicine, Imperial College London, United Kingdom.,Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Lysia Demetriou
- Investigative Medicine, Imperial College London, United Kingdom.,Imanova Centre for Imaging Sciences, Imperial College London, United Kingdom
| | - Matthew B Wall
- Imanova Centre for Imaging Sciences, Imperial College London, United Kingdom.,Division of Brain Sciences, Imperial College London, United Kingdom
| | - Amar J Shah
- Investigative Medicine, Imperial College London, United Kingdom
| | - Sophie A Clarke
- Investigative Medicine, Imperial College London, United Kingdom
| | | | | | | | - Julia K Prague
- Investigative Medicine, Imperial College London, United Kingdom
| | - Ali Abbara
- Investigative Medicine, Imperial College London, United Kingdom
| | | | - Lisa Yang
- Investigative Medicine, Imperial College London, United Kingdom
| | - Victoria Salem
- Investigative Medicine, Imperial College London, United Kingdom
| | | | | | - Mark Tanner
- Imanova Centre for Imaging Sciences, Imperial College London, United Kingdom
| | | | - Amrish Mehta
- Department of Neuroradiology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - John McGonigle
- Imanova Centre for Imaging Sciences, Imperial College London, United Kingdom
| | - Eugenii A Rabiner
- Imanova Centre for Imaging Sciences, Imperial College London, United Kingdom.,Centre for Neuroimaging Sciences, King's College London, United Kingdom
| | - Stephen R Bloom
- Investigative Medicine, Imperial College London, United Kingdom
| | - Waljit S Dhillo
- Investigative Medicine, Imperial College London, United Kingdom
| |
Collapse
|
79
|
Tahedl M, Levine SM, Greenlee MW, Weissert R, Schwarzbach JV. Functional Connectivity in Multiple Sclerosis: Recent Findings and Future Directions. Front Neurol 2018; 9:828. [PMID: 30364281 PMCID: PMC6193088 DOI: 10.3389/fneur.2018.00828] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 09/14/2018] [Indexed: 02/03/2023] Open
Abstract
Multiple sclerosis is a debilitating disorder resulting from scattered lesions in the central nervous system. Because of the high variability of the lesion patterns between patients, it is difficult to relate existing biomarkers to symptoms and their progression. The scattered nature of lesions in multiple sclerosis offers itself to be studied through the lens of network analyses. Recent research into multiple sclerosis has taken such a network approach by making use of functional connectivity. In this review, we briefly introduce measures of functional connectivity and how to compute them. We then identify several common observations resulting from this approach: (a) high likelihood of altered connectivity in deep-gray matter regions, (b) decrease of brain modularity, (c) hemispheric asymmetries in connectivity alterations, and (d) correspondence of behavioral symptoms with task-related and task-unrelated networks. We propose incorporating such connectivity analyses into longitudinal studies in order to improve our understanding of the underlying mechanisms affected by multiple sclerosis, which can consequently offer a promising route to individualizing imaging-related biomarkers for multiple sclerosis.
Collapse
Affiliation(s)
- Marlene Tahedl
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
- Institute for Experimental Psychology, University of Regensburg, Regensburg, Germany
| | - Seth M. Levine
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Mark W. Greenlee
- Institute for Experimental Psychology, University of Regensburg, Regensburg, Germany
| | - Robert Weissert
- Department of Neurology, University of Regensburg, Regensburg, Germany
| | - Jens V. Schwarzbach
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| |
Collapse
|
80
|
Rocca MA, Barkhof F, De Luca J, Frisén J, Geurts JJG, Hulst HE, Sastre-Garriga J, Filippi M. The hippocampus in multiple sclerosis. Lancet Neurol 2018; 17:918-926. [PMID: 30264730 DOI: 10.1016/s1474-4422(18)30309-0] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 07/18/2018] [Accepted: 08/06/2018] [Indexed: 12/16/2022]
Abstract
Some of the clinical manifestations of multiple sclerosis, such as memory impairment and depression, are, at least partly, related to involvement of the hippocampus. Pathological studies have shown extensive demyelination, neuronal damage, and synaptic abnormalities in the hippocampus of patients with multiple sclerosis, and improvements in MRI technology have provided novel ways to assess hippocampal involvement in vivo. It is now accepted that clinical manifestations related to the hippocampus are due not only to focal hippocampal damage, but also to disconnection of the hippocampus from several brain networks. Evidence suggests anatomical and functional subspecialisation of the different hippocampal subfields, resulting in variability between regions in the extent to which damage and repair occur. The hippocampus also has important roles in plasticity and neurogenesis, both of which potentially contribute to functional preservation and restoration. These findings underline the importance of evaluation of the hippocampus not only to improve understanding of the clinical manifestations of multiple sclerosis, but also as a potential future target for treatment.
Collapse
Affiliation(s)
- Maria A Rocca
- Neuroimaging Research Unit and Department of Neurology, Institute of Experimental Neurology, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC-location VUmc, Amsterdam, Netherlands; Institute of Neurology and Institute of Healthcare Engineering, UCL Institute of Neurology, London, UK
| | - John De Luca
- Kessler Foundation, West Orange, NJ, USA; Department of Physical Medicine and Rehabilitation, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Jonas Frisén
- Department of Cell and Molecular Biology, Karolinska Institute, Sweden
| | - Jeroen J G Geurts
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC-location VUmc, Amsterdam, Netherlands
| | - Hanneke E Hulst
- Department of Anatomy and Neurosciences, MS Center Amsterdam, Amsterdam Neuroscience, Amsterdam UMC-location VUmc, Amsterdam, Netherlands
| | - Jaume Sastre-Garriga
- Department of Neurology/Neuroimmunology, Multiple Sclerosis Centre of Catalonia (Cemcat), Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Massimo Filippi
- Neuroimaging Research Unit and Department of Neurology, Institute of Experimental Neurology, San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy.
| | | |
Collapse
|
81
|
Morris G, Reiche EMV, Murru A, Carvalho AF, Maes M, Berk M, Puri BK. Multiple Immune-Inflammatory and Oxidative and Nitrosative Stress Pathways Explain the Frequent Presence of Depression in Multiple Sclerosis. Mol Neurobiol 2018; 55:6282-6306. [PMID: 29294244 PMCID: PMC6061180 DOI: 10.1007/s12035-017-0843-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 12/14/2017] [Indexed: 12/21/2022]
Abstract
Patients with a diagnosis of multiple sclerosis (MS) or major depressive disorder (MDD) share a wide array of biological abnormalities which are increasingly considered to play a contributory role in the pathogenesis and pathophysiology of both illnesses. Shared abnormalities include peripheral inflammation, neuroinflammation, chronic oxidative and nitrosative stress, mitochondrial dysfunction, gut dysbiosis, increased intestinal barrier permeability with bacterial translocation into the systemic circulation, neuroendocrine abnormalities and microglial pathology. Patients with MS and MDD also display a wide range of neuroimaging abnormalities and patients with MS who display symptoms of depression present with different neuroimaging profiles compared with MS patients who are depression-free. The precise details of such pathology are markedly different however. The recruitment of activated encephalitogenic Th17 T cells and subsequent bidirectional interaction leading to classically activated microglia is now considered to lie at the core of MS-specific pathology. The presence of activated microglia is common to both illnesses although the pattern of such action throughout the brain appears to be different. Upregulation of miRNAs also appears to be involved in microglial neurotoxicity and indeed T cell pathology in MS but does not appear to play a major role in MDD. It is suggested that the antidepressant lofepramine, and in particular its active metabolite desipramine, may be beneficial not only for depressive symptomatology but also for the neurological symptoms of MS. One clinical trial has been carried out thus far with, in particular, promising MRI findings.
Collapse
Affiliation(s)
- Gerwyn Morris
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Barwon Health, Geelong, Australia
| | - Edna Maria Vissoci Reiche
- Department of Pathology, Clinical Analysis, and Toxicology, Health Sciences Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Andrea Murru
- Bipolar Disorders Program, Hospital Clínic Barcelona, IDIBAPS, CIBERSAM, Barcelona, Spain
| | - André F Carvalho
- Department of Clinical Medicine and Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceará, Fortaleza, CE, Brazil
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Barwon Health, Geelong, Australia
- Department of Psychiatry, Medical University Plovdiv, Plovdiv, Bulgaria
- Department of Psychiatry, Faculty of Medicine, State University of Londrina, Londrina, Brazil
- Revitalis, Waalre, The Netherlands
- Orygen - The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and the Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, Australia
| | - Basant K Puri
- Department of Medicine, Imperial College London, Hammersmith Hospital, London, UK.
| |
Collapse
|
82
|
Abstract
Multiple sclerosis is a multifactorial disease with heterogeneous pathogenetic mechanisms, which deserve to be studied to evaluate new possible targets for treatments and improve patient management. MR spectroscopy and PET allow assessing in vivo the molecular and metabolic mechanisms underlying the pathogenesis of multiple sclerosis. This article focuses on the relationship between these imaging techniques and the biologic and chemical pathways leading to multiple sclerosis pathology and its clinical features. Future directions of research are also presented.
Collapse
Affiliation(s)
- Marcello Moccia
- NMR Research Unit, Queen Square MS Centre, University College London, Institute of Neurology, 10-12 Russell Square, London WC1B 5EH, UK; MS Clinical Care and Research Centre, Department of Neuroscience, Federico II University, Via Sergio Pansini 5, Naples 80131, Italy
| | - Olga Ciccarelli
- NMR Research Unit, Queen Square MS Centre, University College London, Institute of Neurology, 10-12 Russell Square, London WC1B 5EH, UK; NIHR University College London Hospitals, Biomedical Research Centre, Maple House Suite A 1st floor, 149 Tottenham Court Road, London W1T 7DN, UK.
| |
Collapse
|
83
|
Mohammadyfar MA, Azizpour M, Najafi M, Nooripour R. Comparison of audio-visual short-term and active memory in multiple sclerosis patients and non-patients regarding their depression, stress and anxiety level. NORDIC PSYCHOLOGY 2018; 70:115-128. [DOI: 10.1080/19012276.2017.1362989] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | | | - Mahmood Najafi
- Department of Psychology, Semnan University, Semnan, Iran
| | - Roghieh Nooripour
- Faculty of Education & Psychology, Department of Counseling, Alzahra University, Tehran, Iran
| |
Collapse
|
84
|
Airas L, Nylund M, Rissanen E. Evaluation of Microglial Activation in Multiple Sclerosis Patients Using Positron Emission Tomography. Front Neurol 2018; 9:181. [PMID: 29632509 PMCID: PMC5879102 DOI: 10.3389/fneur.2018.00181] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 03/08/2018] [Indexed: 01/24/2023] Open
Abstract
Understanding the mechanisms underlying progression in multiple sclerosis (MS) is one of the key elements contributing to the identification of appropriate therapeutic targets for this under-managed condition. In addition to plaque-related focal inflammatory pathology typical for relapsing remitting MS there are, in progressive MS, widespread diffuse alterations in brain areas outside the focal lesions. This diffuse pathology is tightly related to microglial activation and is co-localized with signs of neurodegeneration. Microglia are brain-resident cells of the innate immune system and overactivation of microglia is associated with several neurodegenerative diseases. Understanding the role of microglial activation in relation to developing neurodegeneration and disease progression may provide a key to developing therapies to target progressive MS. 18-kDa translocator protein (TSPO) is a mitochondrial molecule upregulated in microglia upon their activation. Positron emission tomography (PET) imaging using TSPO-binding radioligands provides a method to assess microglial activation in patients in vivo. In this mini-review, we summarize the current status of TSPO imaging in the field of MS. In addition, the review discusses new insights into the potential use of this method in treatment trials and in clinical assessment of progressive MS.
Collapse
Affiliation(s)
- Laura Airas
- Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland.,Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Marjo Nylund
- Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland.,Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Eero Rissanen
- Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland.,Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| |
Collapse
|
85
|
Abstract
Depressive disorders are common in patients with multiple sclerosis, influencing their quality of life and adherence to treatments, as well as becoming more frequent with the progression of the disease and in the secondary progressive form of multiple sclerosis. Patients with multiple sclerosis often experience a typical cluster of symptoms in association with depression, such as fatigue, pain and cognitive impairment. However, the pathogenesis of multiple sclerosis-related depression remains partially unclear, even though genetic, immune-inflammatory and psychosocial factors might be seen to play a role, in addition to the brain structural alterations documented by magnetic resonance imaging studies. The high incidence and burden of depression in people affected with multiple sclerosis are matters of crucial importance. Despite such importance, the efficacy of pharmacologic treatments has been poorly studied and, for the most part, the access to non-pharmacological treatments is partially dependent on the local health system availability. It has been determined that interferon-beta and glatiramer acetate do not cause depressive symptoms; however, no definitive data in this regard are avaible for the newer disease-modifyng medications. In this review, we discuss the diagnosis, prevalence, pathogenesis, clinical aspects, magnetic resonance imaging findings and treatments available in patients experiencing multiple sclerosis-related depression.
Collapse
|
86
|
Sumowski JF, Benedict R, Enzinger C, Filippi M, Geurts JJ, Hamalainen P, Hulst H, Inglese M, Leavitt VM, Rocca MA, Rosti-Otajarvi EM, Rao S. Cognition in multiple sclerosis: State of the field and priorities for the future. Neurology 2018; 90:278-288. [PMID: 29343470 PMCID: PMC5818015 DOI: 10.1212/wnl.0000000000004977] [Citation(s) in RCA: 392] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 10/10/2017] [Indexed: 12/15/2022] Open
Abstract
Cognitive decline is recognized as a prevalent and debilitating symptom of multiple sclerosis (MS), especially deficits in episodic memory and processing speed. The field aims to (1) incorporate cognitive assessment into standard clinical care and clinical trials, (2) utilize state-of-the-art neuroimaging to more thoroughly understand neural bases of cognitive deficits, and (3) develop effective, evidence-based, clinically feasible interventions to prevent or treat cognitive dysfunction, which are lacking. There are obstacles to these goals. Our group of MS researchers and clinicians with varied expertise took stock of the current state of the field, and we identify several important practical and theoretical challenges, including key knowledge gaps and methodologic limitations related to (1) understanding and measurement of cognitive deficits, (2) neuroimaging of neural bases and correlates of deficits, and (3) development of effective treatments. This is not a comprehensive review of the extensive literature, but instead a statement of guidelines and priorities for the field. For instance, we provide recommendations for improving the scientific basis and methodologic rigor for cognitive rehabilitation research. Toward this end, we call for multidisciplinary collaborations toward development of biologically based theoretical models of cognition capable of empirical validation and evidence-based refinement, providing the scientific context for effective treatment discovery.
Collapse
Affiliation(s)
- James F Sumowski
- From the Department of Neurology & Corinne Goldsmith Dickinson Center for Multiple Sclerosis (J.F.S., M.I.), Icahn School of Medicine at Mount Sinai, New York; Department of Neurology (R.B.), School of Medicine and Biomedical Sciences, University of Buffalo, State University of New York (SUNY); Department of Neurology (C.E.), Medical University of Graz, Austria; Department of Neurology & Neuroimaging Research Unit, Division of Neuroscience (M.F., M.A.R.), San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy; Department of Anatomy and Neurosciences (J.J.G., H.H.), VU University Medical Center, Amsterdam Neuroscience, VUmc MS Center Amsterdam, the Netherlands; Masku Neurological Rehabilitation Centre (P.H.), Masku, Finland; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (M.I.), University of Genoa, Italy; Department of Neurology & Columbia University Multiple Sclerosis Clinical Care and Research Center (V.M.L.), Columbia University Medical Center, New York, NY; Department of Neurology and Rehabilitation (E.M.R.-O.), Tampere University Hospital, Finland; and Schey Center for Cognitive Neuroimaging, Neurological Institute (S.R.), Cleveland Clinic, OH.
| | - Ralph Benedict
- From the Department of Neurology & Corinne Goldsmith Dickinson Center for Multiple Sclerosis (J.F.S., M.I.), Icahn School of Medicine at Mount Sinai, New York; Department of Neurology (R.B.), School of Medicine and Biomedical Sciences, University of Buffalo, State University of New York (SUNY); Department of Neurology (C.E.), Medical University of Graz, Austria; Department of Neurology & Neuroimaging Research Unit, Division of Neuroscience (M.F., M.A.R.), San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy; Department of Anatomy and Neurosciences (J.J.G., H.H.), VU University Medical Center, Amsterdam Neuroscience, VUmc MS Center Amsterdam, the Netherlands; Masku Neurological Rehabilitation Centre (P.H.), Masku, Finland; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (M.I.), University of Genoa, Italy; Department of Neurology & Columbia University Multiple Sclerosis Clinical Care and Research Center (V.M.L.), Columbia University Medical Center, New York, NY; Department of Neurology and Rehabilitation (E.M.R.-O.), Tampere University Hospital, Finland; and Schey Center for Cognitive Neuroimaging, Neurological Institute (S.R.), Cleveland Clinic, OH
| | - Christian Enzinger
- From the Department of Neurology & Corinne Goldsmith Dickinson Center for Multiple Sclerosis (J.F.S., M.I.), Icahn School of Medicine at Mount Sinai, New York; Department of Neurology (R.B.), School of Medicine and Biomedical Sciences, University of Buffalo, State University of New York (SUNY); Department of Neurology (C.E.), Medical University of Graz, Austria; Department of Neurology & Neuroimaging Research Unit, Division of Neuroscience (M.F., M.A.R.), San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy; Department of Anatomy and Neurosciences (J.J.G., H.H.), VU University Medical Center, Amsterdam Neuroscience, VUmc MS Center Amsterdam, the Netherlands; Masku Neurological Rehabilitation Centre (P.H.), Masku, Finland; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (M.I.), University of Genoa, Italy; Department of Neurology & Columbia University Multiple Sclerosis Clinical Care and Research Center (V.M.L.), Columbia University Medical Center, New York, NY; Department of Neurology and Rehabilitation (E.M.R.-O.), Tampere University Hospital, Finland; and Schey Center for Cognitive Neuroimaging, Neurological Institute (S.R.), Cleveland Clinic, OH
| | - Massimo Filippi
- From the Department of Neurology & Corinne Goldsmith Dickinson Center for Multiple Sclerosis (J.F.S., M.I.), Icahn School of Medicine at Mount Sinai, New York; Department of Neurology (R.B.), School of Medicine and Biomedical Sciences, University of Buffalo, State University of New York (SUNY); Department of Neurology (C.E.), Medical University of Graz, Austria; Department of Neurology & Neuroimaging Research Unit, Division of Neuroscience (M.F., M.A.R.), San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy; Department of Anatomy and Neurosciences (J.J.G., H.H.), VU University Medical Center, Amsterdam Neuroscience, VUmc MS Center Amsterdam, the Netherlands; Masku Neurological Rehabilitation Centre (P.H.), Masku, Finland; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (M.I.), University of Genoa, Italy; Department of Neurology & Columbia University Multiple Sclerosis Clinical Care and Research Center (V.M.L.), Columbia University Medical Center, New York, NY; Department of Neurology and Rehabilitation (E.M.R.-O.), Tampere University Hospital, Finland; and Schey Center for Cognitive Neuroimaging, Neurological Institute (S.R.), Cleveland Clinic, OH
| | - Jeroen J Geurts
- From the Department of Neurology & Corinne Goldsmith Dickinson Center for Multiple Sclerosis (J.F.S., M.I.), Icahn School of Medicine at Mount Sinai, New York; Department of Neurology (R.B.), School of Medicine and Biomedical Sciences, University of Buffalo, State University of New York (SUNY); Department of Neurology (C.E.), Medical University of Graz, Austria; Department of Neurology & Neuroimaging Research Unit, Division of Neuroscience (M.F., M.A.R.), San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy; Department of Anatomy and Neurosciences (J.J.G., H.H.), VU University Medical Center, Amsterdam Neuroscience, VUmc MS Center Amsterdam, the Netherlands; Masku Neurological Rehabilitation Centre (P.H.), Masku, Finland; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (M.I.), University of Genoa, Italy; Department of Neurology & Columbia University Multiple Sclerosis Clinical Care and Research Center (V.M.L.), Columbia University Medical Center, New York, NY; Department of Neurology and Rehabilitation (E.M.R.-O.), Tampere University Hospital, Finland; and Schey Center for Cognitive Neuroimaging, Neurological Institute (S.R.), Cleveland Clinic, OH
| | - Paivi Hamalainen
- From the Department of Neurology & Corinne Goldsmith Dickinson Center for Multiple Sclerosis (J.F.S., M.I.), Icahn School of Medicine at Mount Sinai, New York; Department of Neurology (R.B.), School of Medicine and Biomedical Sciences, University of Buffalo, State University of New York (SUNY); Department of Neurology (C.E.), Medical University of Graz, Austria; Department of Neurology & Neuroimaging Research Unit, Division of Neuroscience (M.F., M.A.R.), San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy; Department of Anatomy and Neurosciences (J.J.G., H.H.), VU University Medical Center, Amsterdam Neuroscience, VUmc MS Center Amsterdam, the Netherlands; Masku Neurological Rehabilitation Centre (P.H.), Masku, Finland; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (M.I.), University of Genoa, Italy; Department of Neurology & Columbia University Multiple Sclerosis Clinical Care and Research Center (V.M.L.), Columbia University Medical Center, New York, NY; Department of Neurology and Rehabilitation (E.M.R.-O.), Tampere University Hospital, Finland; and Schey Center for Cognitive Neuroimaging, Neurological Institute (S.R.), Cleveland Clinic, OH
| | - Hanneke Hulst
- From the Department of Neurology & Corinne Goldsmith Dickinson Center for Multiple Sclerosis (J.F.S., M.I.), Icahn School of Medicine at Mount Sinai, New York; Department of Neurology (R.B.), School of Medicine and Biomedical Sciences, University of Buffalo, State University of New York (SUNY); Department of Neurology (C.E.), Medical University of Graz, Austria; Department of Neurology & Neuroimaging Research Unit, Division of Neuroscience (M.F., M.A.R.), San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy; Department of Anatomy and Neurosciences (J.J.G., H.H.), VU University Medical Center, Amsterdam Neuroscience, VUmc MS Center Amsterdam, the Netherlands; Masku Neurological Rehabilitation Centre (P.H.), Masku, Finland; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (M.I.), University of Genoa, Italy; Department of Neurology & Columbia University Multiple Sclerosis Clinical Care and Research Center (V.M.L.), Columbia University Medical Center, New York, NY; Department of Neurology and Rehabilitation (E.M.R.-O.), Tampere University Hospital, Finland; and Schey Center for Cognitive Neuroimaging, Neurological Institute (S.R.), Cleveland Clinic, OH
| | - Matilde Inglese
- From the Department of Neurology & Corinne Goldsmith Dickinson Center for Multiple Sclerosis (J.F.S., M.I.), Icahn School of Medicine at Mount Sinai, New York; Department of Neurology (R.B.), School of Medicine and Biomedical Sciences, University of Buffalo, State University of New York (SUNY); Department of Neurology (C.E.), Medical University of Graz, Austria; Department of Neurology & Neuroimaging Research Unit, Division of Neuroscience (M.F., M.A.R.), San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy; Department of Anatomy and Neurosciences (J.J.G., H.H.), VU University Medical Center, Amsterdam Neuroscience, VUmc MS Center Amsterdam, the Netherlands; Masku Neurological Rehabilitation Centre (P.H.), Masku, Finland; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (M.I.), University of Genoa, Italy; Department of Neurology & Columbia University Multiple Sclerosis Clinical Care and Research Center (V.M.L.), Columbia University Medical Center, New York, NY; Department of Neurology and Rehabilitation (E.M.R.-O.), Tampere University Hospital, Finland; and Schey Center for Cognitive Neuroimaging, Neurological Institute (S.R.), Cleveland Clinic, OH
| | - Victoria M Leavitt
- From the Department of Neurology & Corinne Goldsmith Dickinson Center for Multiple Sclerosis (J.F.S., M.I.), Icahn School of Medicine at Mount Sinai, New York; Department of Neurology (R.B.), School of Medicine and Biomedical Sciences, University of Buffalo, State University of New York (SUNY); Department of Neurology (C.E.), Medical University of Graz, Austria; Department of Neurology & Neuroimaging Research Unit, Division of Neuroscience (M.F., M.A.R.), San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy; Department of Anatomy and Neurosciences (J.J.G., H.H.), VU University Medical Center, Amsterdam Neuroscience, VUmc MS Center Amsterdam, the Netherlands; Masku Neurological Rehabilitation Centre (P.H.), Masku, Finland; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (M.I.), University of Genoa, Italy; Department of Neurology & Columbia University Multiple Sclerosis Clinical Care and Research Center (V.M.L.), Columbia University Medical Center, New York, NY; Department of Neurology and Rehabilitation (E.M.R.-O.), Tampere University Hospital, Finland; and Schey Center for Cognitive Neuroimaging, Neurological Institute (S.R.), Cleveland Clinic, OH
| | - Maria A Rocca
- From the Department of Neurology & Corinne Goldsmith Dickinson Center for Multiple Sclerosis (J.F.S., M.I.), Icahn School of Medicine at Mount Sinai, New York; Department of Neurology (R.B.), School of Medicine and Biomedical Sciences, University of Buffalo, State University of New York (SUNY); Department of Neurology (C.E.), Medical University of Graz, Austria; Department of Neurology & Neuroimaging Research Unit, Division of Neuroscience (M.F., M.A.R.), San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy; Department of Anatomy and Neurosciences (J.J.G., H.H.), VU University Medical Center, Amsterdam Neuroscience, VUmc MS Center Amsterdam, the Netherlands; Masku Neurological Rehabilitation Centre (P.H.), Masku, Finland; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (M.I.), University of Genoa, Italy; Department of Neurology & Columbia University Multiple Sclerosis Clinical Care and Research Center (V.M.L.), Columbia University Medical Center, New York, NY; Department of Neurology and Rehabilitation (E.M.R.-O.), Tampere University Hospital, Finland; and Schey Center for Cognitive Neuroimaging, Neurological Institute (S.R.), Cleveland Clinic, OH
| | - Eija M Rosti-Otajarvi
- From the Department of Neurology & Corinne Goldsmith Dickinson Center for Multiple Sclerosis (J.F.S., M.I.), Icahn School of Medicine at Mount Sinai, New York; Department of Neurology (R.B.), School of Medicine and Biomedical Sciences, University of Buffalo, State University of New York (SUNY); Department of Neurology (C.E.), Medical University of Graz, Austria; Department of Neurology & Neuroimaging Research Unit, Division of Neuroscience (M.F., M.A.R.), San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy; Department of Anatomy and Neurosciences (J.J.G., H.H.), VU University Medical Center, Amsterdam Neuroscience, VUmc MS Center Amsterdam, the Netherlands; Masku Neurological Rehabilitation Centre (P.H.), Masku, Finland; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (M.I.), University of Genoa, Italy; Department of Neurology & Columbia University Multiple Sclerosis Clinical Care and Research Center (V.M.L.), Columbia University Medical Center, New York, NY; Department of Neurology and Rehabilitation (E.M.R.-O.), Tampere University Hospital, Finland; and Schey Center for Cognitive Neuroimaging, Neurological Institute (S.R.), Cleveland Clinic, OH
| | - Stephen Rao
- From the Department of Neurology & Corinne Goldsmith Dickinson Center for Multiple Sclerosis (J.F.S., M.I.), Icahn School of Medicine at Mount Sinai, New York; Department of Neurology (R.B.), School of Medicine and Biomedical Sciences, University of Buffalo, State University of New York (SUNY); Department of Neurology (C.E.), Medical University of Graz, Austria; Department of Neurology & Neuroimaging Research Unit, Division of Neuroscience (M.F., M.A.R.), San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy; Department of Anatomy and Neurosciences (J.J.G., H.H.), VU University Medical Center, Amsterdam Neuroscience, VUmc MS Center Amsterdam, the Netherlands; Masku Neurological Rehabilitation Centre (P.H.), Masku, Finland; Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (M.I.), University of Genoa, Italy; Department of Neurology & Columbia University Multiple Sclerosis Clinical Care and Research Center (V.M.L.), Columbia University Medical Center, New York, NY; Department of Neurology and Rehabilitation (E.M.R.-O.), Tampere University Hospital, Finland; and Schey Center for Cognitive Neuroimaging, Neurological Institute (S.R.), Cleveland Clinic, OH
| |
Collapse
|
87
|
Notter T, Coughlin JM, Sawa A, Meyer U. Reconceptualization of translocator protein as a biomarker of neuroinflammation in psychiatry. Mol Psychiatry 2018; 23:36-47. [PMID: 29203847 DOI: 10.1038/mp.2017.232] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/05/2017] [Accepted: 10/02/2017] [Indexed: 02/06/2023]
Abstract
A great deal of interest in psychiatric research is currently centered upon the pathogenic role of inflammatory processes. Positron emission tomography (PET) using radiolabeled ligands selective for the 18 kDa translocator protein (TSPO) has become the most widely used technique to assess putative neuroimmune abnormalities in vivo. Originally used to detect discrete neurotoxic damages, TSPO has generally turned into a biomarker of 'neuroinflammation' or 'microglial activation'. Psychiatric research has mostly accepted these denotations of TSPO, even if they may be inadequate and misleading under many pathological conditions. A reliable and neurobiologically meaningful diagnosis of 'neuroinflammation' or 'microglial activation' is unlikely to be achieved by the sole use of TSPO PET imaging. It is also very likely that the pathological meanings of altered TSPO binding or expression are disease-specific, and therefore, not easily generalizable across different neuropathologies or inflammatory conditions. This difficulty is intricately linked to the varying (and still ill-defined) physiological functions and cellular expression patterns of TSPO in health and disease. While altered TSPO binding or expression may indeed mirror ongoing neuroinflammatory processes in some cases, it may reflect other pathophysiological processes such as abnormalities in cell metabolism, energy production and oxidative stress in others. Hence, the increasing popularity of TSPO PET imaging has paradoxically introduced substantial uncertainty regarding the nature and meaning of neuroinflammatory processes and microglial activation in psychiatry, and likely in other neuropathological conditions as well. The ambiguity of conceiving TSPO simply as a biomarker of 'neuroinflammation' or 'microglial activation' calls for alternative interpretations and complimentary approaches. Without the latter, the ongoing scientific efforts and excitement surrounding the role of the neuroimmune system in psychiatry may not turn into therapeutic hope for affected individuals.
Collapse
Affiliation(s)
- T Notter
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - J M Coughlin
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, MD, USA.,Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - A Sawa
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - U Meyer
- Institute of Pharmacology and Toxicology, University of Zurich-Vetsuisse, Zurich, Switzerland.,Neuroscience Center Zurich, University of Zurich and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
88
|
Abstract
Depressive disorders occur in up to 50% of people living with multiple sclerosis (MS). Prevalence estimates are generally 2-3-times higher than those of the general population. Myriad aetiologic factors may contribute to the aetiology of depression in MS including biological mechanisms (e.g. hippocampal microglial activation, lesion burden, regional atrophy), as well as the stressors, threats, and losses that accompany living with an unpredictable and often disabling disease. Some prominent risk factors for depression such as (younger) age, (female) sex, and family history of depression are less consistently associated with depression in MS than they are in the general population. Management of depression in MS has not been well studied, but available data on detection and treatment align with general principles of depression management. While the validity of standard measurement scales has often been questioned, available evidence suggests that standard scales provide valid ratings. Evidence for the effectiveness of depression treatments in MS is limited, but available evidence supports the effectiveness of standard treatment approaches, including both cognitive behavioural therapies and antidepressant medications.
Collapse
Affiliation(s)
- Scott B Patten
- a Department of Community Health Sciences , University of Calgary, Calgary , Alberta , Canada
| | - Ruth Ann Marrie
- b Department of Internal Medicine (Neurology) , University of Manitoba , Manitoba , Canada.,c Department of Community Health Sciences , University of Manitoba , Manitoba , Canada
| | - Mauro G Carta
- d Department of Medical Sciences and Public Health , Quality of Care, University of Cagliari , Cagliari , Italy
| |
Collapse
|
89
|
Ayatollahi AM, Haji Molla Hoseini M, Ghanadian SM, Kosari-Nasab M, Mami F, Yazdiniapoure Z, Zolfaghari B, Salari AA. TAMEC: a new analogue of cyclomyrsinol diterpenes decreases anxiety- and depression-like behaviors in a mouse model of multiple sclerosis. Neurol Res 2017; 39:1056-1065. [DOI: 10.1080/01616412.2017.1376789] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Abdul Majid Ayatollahi
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- School of Pharmacy, Department of Pharmacognosy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Haji Molla Hoseini
- Faculty of Medicine, Department of Immunology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Syed Mustafa Ghanadian
- Pharmaceutical Sciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Morteza Kosari-Nasab
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Mami
- Faculty of Medicine, Department of Immunology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeinab Yazdiniapoure
- Pharmaceutical Sciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Behzad Zolfaghari
- Pharmaceutical Sciences Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali-Akbar Salari
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Salari Institute of Cognitive and Behavioral Disorders, Alborz, Iran
| |
Collapse
|
90
|
Rossi S, Studer V, Motta C, Polidoro S, Perugini J, Macchiarulo G, Giovannetti AM, Pareja-Gutierrez L, Calò A, Colonna I, Furlan R, Martino G, Centonze D. Neuroinflammation drives anxiety and depression in relapsing-remitting multiple sclerosis. Neurology 2017; 89:1338-1347. [DOI: 10.1212/wnl.0000000000004411] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 06/13/2017] [Indexed: 11/15/2022] Open
Abstract
Objective:To explore the inflammatory processes in the pathogenesis of psychiatric symptoms and the prognostic value of psychiatric comorbidities in multiple sclerosis (MS).Methods:Four hundred five patients with relapsing-remitting (RR) MS underwent psychiatric evaluation by means of Beck Depression Inventory II (BDI-II) and State/Trait Anxiety Inventory (STAI-Y). The inflammatory activity level was assessed by MRI. In a subset of 111 treatment-naive patients, CSF levels of proinflammatory cytokines were determined. Correlation and regression analyses were performed to determine associations between variables.Results:Relapsing patients demonstrated greater values of STAI-state and BDI-II compared with remitting patients but comparable trait-anxiety scores. There were no significant differences in psychometric parameters between relapsing and asymptomatic MRI-active patients, highlighting the effect of subclinical inflammation on mood disturbances. A significant reduction of STAI-state and BDI-II scores was recorded, along with the subsiding of neuroinflammation. Interleukin-2 CSF levels were found to correlate with STAI-state, while tumor necrosis factor-α and interleukin-1β correlated with BDI-II. Because emotional disorders were associated with subclinical inflammation, variations of the psychometric profile were able to detect subclinical reactivation earlier. In line with this, high STAI-state values considerably predicted the possibility of disease reactivation.Conclusions:Mood alterations are induced by intrathecal inflammation, even though not clinically apparent, and are able to predict inflammatory reactivations in RRMS. Inflammation is therefore a biological event, not less important than the traditional psychosocial factors, involved in mood disorders.
Collapse
|
91
|
Effect of Cigarette Smoking on a Marker for Neuroinflammation: A [ 11C]DAA1106 Positron Emission Tomography Study. Neuropsychopharmacology 2017; 42:1630-1639. [PMID: 28262740 PMCID: PMC5518907 DOI: 10.1038/npp.2017.48] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Revised: 02/01/2017] [Accepted: 02/23/2017] [Indexed: 01/21/2023]
Abstract
In the brain, microglia continuously scan the surrounding extracellular space in order to respond to damage or infection by becoming activated and participating in neuroinflammation. When activated, microglia increase the expression of translocator protein (TSPO) 18 kDa, thereby making the TSPO expression a marker for neuroinflammation. We used the radiotracer [11C]DAA1106 (a ligand for TSPO) and positron emission tomography (PET) to determine the effect of smoking on availability of this marker for neuroinflammation. Forty-five participants (30 smokers and 15 non-smokers) completed the study and had usable data. Participants underwent a dynamic PET scanning session with bolus injection of [11C]DAA1106 (with smokers in the satiated state) and blood draws during PET scanning to determine TSPO affinity genotype and plasma nicotine levels. Whole-brain standardized uptake values (SUVs) were determined, and analysis of variance was performed, with group (smoker vs non-smoker) and genotype as factors, thereby controlling for genotype. Smokers and non-smokers differed in whole-brain SUVs (P=0.006) owing to smokers having 16.8% lower values than non-smokers. The groups did not differ in injected radiotracer dose or body weight, which were used to calculate SUV. An inverse association was found between whole-brain SUV and reported cigarettes per day (P<0.05), but no significant relationship was found for plasma nicotine. Thus, smokers have less [11C]DAA1106 binding globally than non-smokers, indicating less microglial activation. Study findings are consistent with much prior research demonstrating that smokers have impaired inflammatory functioning compared with non-smokers and that constituents of tobacco smoke other than nicotine affect inflammatory processes.
Collapse
|
92
|
A Comparison of Neuroimaging Abnormalities in Multiple Sclerosis, Major Depression and Chronic Fatigue Syndrome (Myalgic Encephalomyelitis): is There a Common Cause? Mol Neurobiol 2017; 55:3592-3609. [PMID: 28516431 PMCID: PMC5842501 DOI: 10.1007/s12035-017-0598-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/03/2017] [Indexed: 01/23/2023]
Abstract
There is copious evidence of abnormalities in resting-state functional network connectivity states, grey and white matter pathology and impaired cerebral perfusion in patients afforded a diagnosis of multiple sclerosis, major depression or chronic fatigue syndrome (CFS) (myalgic encephalomyelitis). Systemic inflammation may well be a major element explaining such findings. Inter-patient and inter-illness variations in neuroimaging findings may arise at least in part from regional genetic, epigenetic and environmental variations in the functions of microglia and astrocytes. Regional differences in neuronal resistance to oxidative and inflammatory insults and in the performance of antioxidant defences in the central nervous system may also play a role. Importantly, replicated experimental findings suggest that the use of high-resolution SPECT imaging may have the capacity to differentiate patients afforded a diagnosis of CFS from those with a diagnosis of depression. Further research involving this form of neuroimaging appears warranted in an attempt to overcome the problem of aetiologically heterogeneous cohorts which probably explain conflicting findings produced by investigative teams active in this field. However, the ionising radiation and relative lack of sensitivity involved probably preclude its use as a routine diagnostic tool.
Collapse
|
93
|
Dupont AC, Largeau B, Santiago Ribeiro MJ, Guilloteau D, Tronel C, Arlicot N. Translocator Protein-18 kDa (TSPO) Positron Emission Tomography (PET) Imaging and Its Clinical Impact in Neurodegenerative Diseases. Int J Mol Sci 2017; 18:ijms18040785. [PMID: 28387722 PMCID: PMC5412369 DOI: 10.3390/ijms18040785] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/31/2017] [Accepted: 04/04/2017] [Indexed: 02/06/2023] Open
Abstract
In vivo exploration of activated microglia in neurodegenerative diseases is achievable by Positron Emission Tomography (PET) imaging, using dedicated radiopharmaceuticals targeting the translocator protein-18 kDa (TSPO). In this review, we emphasized the major advances made over the last 20 years, thanks to TSPO PET imaging, to define the pathophysiological implication of microglia activation and neuroinflammation in neurodegenerative diseases, including Parkinson’s disease, Huntington’s disease, dementia, amyotrophic lateral sclerosis, multiple sclerosis, and also in psychiatric disorders. The extent and upregulation of TSPO as a molecular biomarker of activated microglia in the human brain is now widely documented in these pathologies, but its significance, and especially its protective or deleterious action regarding the disease’s stage, remains under debate. Thus, we exposed new and plausible suggestions to enhance the contribution of TSPO PET imaging for biomedical research by exploring microglia’s role and interactions with other cells in brain parenchyma. Multiplex approaches, associating TSPO PET radiopharmaceuticals with other biomarkers (PET imaging of cellular metabolism, neurotransmission or abnormal protein aggregates, but also other imaging modalities, and peripheral cytokine levels measurement and/or metabolomics analysis) was considered. Finally, the actual clinical impact of TSPO PET imaging as a routine biomarker of neuroinflammation was put into perspective regarding the current development of diagnostic and therapeutic strategies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Anne-Claire Dupont
- CHRU Tours, 2 Boulevard Tonnellé, 37044 Tours, France.
- Institut National de la Santé et de la Recherche Médicale U930, 10 Boulevard Tonnellé, 37032 Tours, France.
| | | | - Maria Joao Santiago Ribeiro
- CHRU Tours, 2 Boulevard Tonnellé, 37044 Tours, France.
- Institut National de la Santé et de la Recherche Médicale U930, 10 Boulevard Tonnellé, 37032 Tours, France.
| | - Denis Guilloteau
- CHRU Tours, 2 Boulevard Tonnellé, 37044 Tours, France.
- Institut National de la Santé et de la Recherche Médicale U930, 10 Boulevard Tonnellé, 37032 Tours, France.
| | - Claire Tronel
- Institut National de la Santé et de la Recherche Médicale U930, 10 Boulevard Tonnellé, 37032 Tours, France.
| | - Nicolas Arlicot
- CHRU Tours, 2 Boulevard Tonnellé, 37044 Tours, France.
- Institut National de la Santé et de la Recherche Médicale U930, 10 Boulevard Tonnellé, 37032 Tours, France.
| |
Collapse
|
94
|
Datta G, Colasanti A, Kalk N, Owen D, Scott G, Rabiner EA, Gunn RN, Lingford-Hughes A, Malik O, Ciccarelli O, Nicholas R, Nei L, Battaglini M, Stefano ND, Matthews PM. 11C-PBR28 and 18F-PBR111 Detect White Matter Inflammatory Heterogeneity in Multiple Sclerosis. J Nucl Med 2017; 58:1477-1482. [DOI: 10.2967/jnumed.116.187161] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 02/28/2017] [Indexed: 11/16/2022] Open
|
95
|
Analysis of ageing-associated grey matter volume in patients with multiple sclerosis shows excess atrophy in subcortical regions. NEUROIMAGE-CLINICAL 2016; 13:9-15. [PMID: 27896065 PMCID: PMC5121150 DOI: 10.1016/j.nicl.2016.11.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 10/05/2016] [Accepted: 11/04/2016] [Indexed: 12/28/2022]
Abstract
Age of onset in multiple sclerosis (MS) exerts an influence on the course of disease. This study examined whether global and regional brain volumes differed between "younger" and "older" onset MS subjects who were matched for short disease duration, mean 1.9 years and burden as measured by the MS Severity Score and relapses. 21 younger-onset MS subjects (age 30.4 ± 3.2 years) were compared with 17 older-onset (age 48.7 ± 3.3 years) as well as age-matched controls (n = 31, 31.9 ± 3.5 years and n = 21, 47.3 ± 4.0 years). All subjects underwent 3D volumetric T1 and T2-FLAIR imaging. White matter (WM) and grey matter (GM) lesions were outlined manually. Lesions were filled prior to tissue and structural segmentation to reduce classification errors. Volume loss versus control was predominantly in the subcortical GM, at > 13% loss. Younger and older-onset MS subjects had similar, strong excess loss in the putamen, thalamus, and nucleus accumbens. No excess loss was detected in the amygdala or pallidum. The hippocampus and caudate showed significant excess loss in the younger group (p < 0.001) and a strong trend in the older-onset group. These results provide a potential imaging correlate of published neuropsychological studies that reported the association of younger age at disease onset with impaired cognitive performance, including decreased working memory.
Collapse
|
96
|
Tyrtyshnaia AA, Lysenko LV, Madamba F, Manzhulo IV, Khotimchenko MY, Kleschevnikov AM. Acute neuroinflammation provokes intracellular acidification in mouse hippocampus. J Neuroinflammation 2016; 13:283. [PMID: 27809864 PMCID: PMC5094044 DOI: 10.1186/s12974-016-0747-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 10/16/2016] [Indexed: 11/10/2022] Open
Abstract
Background Maintaining pH levels within the physiological norm is an important component of brain homeostasis. However, in some pathological or physiological conditions, the capacity of the pH regulatory system could be overpowered by various factors resulting in a transient or permanent alteration in pH levels. Such changes are often observed in pathological conditions associated with neuroinflammation. We hypothesized that neuroinflammation itself is a factor affecting pH levels in neural tissue. To assess this hypothesis, we examined the effects of acute LPS-induced neuroinflammation on intra- and extracellular pH (pHi and pHo) levels in the CA1 region of mouse hippocampus. Methods Acute neuroinflammation was induced using two approaches: (1) in vivo by i.p. injections of LPS (5 mg/kg) and (2) in vitro by incubating hippocampal slices of naïve animals in the LPS-containing media (1 μg/mL, 1 h at 35 °C). Standard techniques were used to prepare hippocampal slices. pHi was measured using ratiometric pH-sensitive fluorescent dye BCECF-AM. pHo was assessed using calibrated pH-sensitive micropipettes. The presence of neuroinflammation was verified with immunohistochemistry (IL-1β and Iba1) and ELISA (IL-1β and TNF-α). Results A significant reduction of pHi was observed in the slices of the LPS-injected 3-month-old (LPS 7.13 ± 0.03; Sal 7.22 ± 0.03; p = 0.043, r = 0.43) and 19-month-old (LPS 6.78 ± 0.08; Sal 7.13 ± 0.03; p = 0.0001, r = 0.32) mice. In contrast, the levels of pHo within the slice, measured in 19-month-old animals, were not affected (LPS 7.27 ± 0.02; Sal 7.26 ± 0.02; p = 0.6, r = 0.13). A reduction of pHi was also observed in the LPS-treated slices during the interval 3.5–7 h after the LPS exposure (LPS 6.92 ± 0.07; Veh 7.28 ± 0.05; p = 0.0001, r = 0.46). Conclusions Acute LPS-induced neuroinflammation results in a significant intracellular acidification of the CA1 neurons in mouse hippocampus, while the pHo remains largely unchanged. Such changes may represent a specific protective reaction of neural tissue in unfavorable external conditions or be a part of the pathological process. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0747-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anna A Tyrtyshnaia
- Department of Neurosciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.,School of Biomedicine, Far Eastern Federal University, Sukhanova 8, Vladivostok, 690950, Russian Federation
| | - Larisa V Lysenko
- Department of Neurosciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.,Academy of Biology and Biotechnology of Southern Federal University, 194/1 Stachki Str, Rostov-na-Donu, 344090, Russian Federation
| | - Francisco Madamba
- Department of Neurosciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Igor V Manzhulo
- School of Biomedicine, Far Eastern Federal University, Sukhanova 8, Vladivostok, 690950, Russian Federation
| | - Maxim Y Khotimchenko
- School of Biomedicine, Far Eastern Federal University, Sukhanova 8, Vladivostok, 690950, Russian Federation
| | - Alexander M Kleschevnikov
- Department of Neurosciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA. .,School of Biomedicine, Far Eastern Federal University, Sukhanova 8, Vladivostok, 690950, Russian Federation.
| |
Collapse
|
97
|
Abstract
The discovery and development of central nervous system (CNS) drugs is an extremely challenging process requiring large resources, timelines, and associated costs. The high risk of failure leads to high levels of risk. Over the past couple of decades PET imaging has become a central component of the CNS drug-development process, enabling decision-making in phase I studies, where early discharge of risk provides increased confidence to progress a candidate to more costly later phase testing at the right dose level or alternatively to kill a compound through failure to meet key criteria. The so called "3 pillars" of drug survival, namely; tissue exposure, target engagement, and pharmacologic activity, are particularly well suited for evaluation by PET imaging. This review introduces the process of CNS drug development before considering how PET imaging of the "3 pillars" has advanced to provide valuable tools for decision-making on the critical path of CNS drug development. Finally, we review the advances in PET science of biomarker development and analysis that enable sophisticated drug-development studies in man.
Collapse
Affiliation(s)
- Roger N Gunn
- Imanova Ltd, London, United Kingdom; Division of Brain Sciences, Imperial College London, London, United Kingdom; Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom.
| | - Eugenii A Rabiner
- Imanova Ltd, London, United Kingdom; Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, United Kingdom
| |
Collapse
|
98
|
Herranz E, Giannì C, Louapre C, Treaba CA, Govindarajan ST, Ouellette R, Loggia ML, Sloane JA, Madigan N, Izquierdo-Garcia D, Ward N, Mangeat G, Granberg T, Klawiter EC, Catana C, Hooker JM, Taylor N, Ionete C, Kinkel RP, Mainero C. Neuroinflammatory component of gray matter pathology in multiple sclerosis. Ann Neurol 2016; 80:776-790. [PMID: 27686563 DOI: 10.1002/ana.24791] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 09/07/2016] [Accepted: 09/25/2016] [Indexed: 12/12/2022]
Abstract
OBJECTIVE In multiple sclerosis (MS), using simultaneous magnetic resonance-positron emission tomography (MR-PET) imaging with 11 C-PBR28, we quantified expression of the 18kDa translocator protein (TSPO), a marker of activated microglia/macrophages, in cortex, cortical lesions, deep gray matter (GM), white matter (WM) lesions, and normal-appearing WM (NAWM) to investigate the in vivo pathological and clinical relevance of neuroinflammation. METHODS Fifteen secondary-progressive MS (SPMS) patients, 12 relapsing-remitting MS (RRMS) patients, and 14 matched healthy controls underwent 11 C-PBR28 MR-PET. MS subjects underwent 7T T2*-weighted imaging for cortical lesion segmentation, and neurological and cognitive evaluation. 11 C-PBR28 binding was measured using normalized 60- to 90-minute standardized uptake values and volume of distribution ratios. RESULTS Relative to controls, MS subjects exhibited abnormally high 11 C-PBR28 binding across the brain, the greatest increases being in cortex and cortical lesions, thalamus, hippocampus, and NAWM. MS WM lesions showed relatively modest TSPO increases. With the exception of cortical lesions, where TSPO expression was similar, 11 C-PBR28 uptake across the brain was greater in SPMS than in RRMS. In MS, increased 11 C-PBR28 binding in cortex, deep GM, and NAWM correlated with neurological disability and impaired cognitive performance; cortical thinning correlated with increased thalamic TSPO levels. INTERPRETATION In MS, neuroinflammation is present in the cortex, cortical lesions, deep GM, and NAWM, is closely linked to poor clinical outcome, and is at least partly linked to neurodegeneration. Distinct inflammatory-mediated factors may underlie accumulation of cortical and WM lesions. Quantification of TSPO levels in MS could prove to be a sensitive tool for evaluating in vivo the inflammatory component of GM pathology, particularly in cortical lesions. Ann Neurol 2016;80:776-790.
Collapse
Affiliation(s)
- Elena Herranz
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA.,Harvard Medical School, Boston, MA
| | - Costanza Giannì
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA.,Harvard Medical School, Boston, MA
| | - Céline Louapre
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA.,Harvard Medical School, Boston, MA
| | - Constantina A Treaba
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA.,Harvard Medical School, Boston, MA
| | - Sindhuja T Govindarajan
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Russell Ouellette
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Marco L Loggia
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA.,Harvard Medical School, Boston, MA
| | - Jacob A Sloane
- Harvard Medical School, Boston, MA.,Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA
| | - Nancy Madigan
- Harvard Medical School, Boston, MA.,Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA
| | - David Izquierdo-Garcia
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA.,Harvard Medical School, Boston, MA
| | - Noreen Ward
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA
| | - Gabriel Mangeat
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA.,Institute of Biomedical Engineering, Polytechnique Montréal, Montréal, QC, Canada
| | - Tobias Granberg
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA.,Harvard Medical School, Boston, MA
| | - Eric C Klawiter
- Harvard Medical School, Boston, MA.,Department of Neurology, Massachusetts General Hospital, Boston, MA
| | - Ciprian Catana
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA.,Harvard Medical School, Boston, MA
| | - Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA.,Harvard Medical School, Boston, MA
| | - Norman Taylor
- Harvard Medical School, Boston, MA.,Department of Anesthesiology, Massachusetts General Hospital, Boston, MA
| | | | - Revere P Kinkel
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA.,University of San Diego, San Diego, California
| | - Caterina Mainero
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA.,Harvard Medical School, Boston, MA
| |
Collapse
|
99
|
Airas L, Rissanen E, Rinne J. Imaging of microglial activation in MS using PET: Research use and potential future clinical application. Mult Scler 2016; 23:496-504. [PMID: 27760860 DOI: 10.1177/1352458516674568] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Multiple sclerosis (MS) is a complex disease, where several processes can be selected as a target for positron emission topography (PET) imaging. Unlike magnetic resonance imaging (MRI), PET provides specific and quantitative information, and unlike neuropathology, it can be non-invasively applied to living patients, which enables longitudinal follow-up of the MS pathology. In the study of MS, PET can be useful for in vivo evaluation of specific pathological characteristics at various stages of the disease. Increased understanding of the progressive MS pathology will enhance the treatment options of this undertreated condition. The ultimate goal of developing and expanding PET in the study of MS is to have clinical non-invasive in vivo imaging biomarkers of neuroinflammation that will help to establish prognosis and accurately measure response to therapeutics. This topical review provides an overview of the promises and challenges of the use of PET in MS.
Collapse
Affiliation(s)
- Laura Airas
- Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland; Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Eero Rissanen
- Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland; Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Juha Rinne
- Division of Clinical Neurosciences, Turku University Hospital and University of Turku, Turku, Finland; Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| |
Collapse
|
100
|
Wall MB, Birch D, Yong MY. Opportunities and considerations for visualising neuroimaging data on very large displays. F1000Res 2016; 5:2157. [PMID: 27703670 PMCID: PMC5031127 DOI: 10.12688/f1000research.9522.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/30/2016] [Indexed: 11/20/2022] Open
Abstract
Neuroimaging experiments can generate impressive volumes of data and many images of the results. This is particularly true of multi-modal imaging studies that use more than one imaging technique, or when imaging is combined with other assessments. A challenge for these studies is appropriate visualisation of results in order to drive insights and guide accurate interpretations. Next-generation visualisation technology therefore has much to offer the neuroimaging community. One example is the Imperial College London Data Observatory; a high-resolution (132 megapixel) arrangement of 64 monitors, arranged in a 313 degree arc, with a 6 metre diameter, powered by 32 rendering nodes. This system has the potential for high-resolution, large-scale display of disparate data types in a space designed to promote collaborative discussion by multiple researchers and/or clinicians. Opportunities for the use of the Data Observatory are discussed, with particular reference to applications in Multiple Sclerosis (MS) research and clinical practice. Technical issues and current work designed to optimise the use of the Data Observatory for neuroimaging are also discussed, as well as possible future research that could be enabled by the use of the system in combination with eye-tracking technology.
Collapse
Affiliation(s)
- Matthew B Wall
- Imanova Centre for Imaging Sciences, London, W12 0NN, UK; Division of Brain Sciences, Imperial College London, London, SW7 2AZ, UK; Clinical Psychopharmacology Unit, University College London, London, WC1E 7HB, UK
| | - David Birch
- Data Science Institute, Imperial College London, London, SW7 2AZ, UK
| | - May Y Yong
- Data Science Institute, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|