1
|
De Simone G, Iasevoli F, Barone A, Gaudieri V, Cuocolo A, Ciccarelli M, Pappatà S, de Bartolomeis A. Addressing brain metabolic connectivity in treatment-resistant schizophrenia: a novel graph theory-driven application of 18F-FDG-PET with antipsychotic dose correction. SCHIZOPHRENIA (HEIDELBERG, GERMANY) 2024; 10:116. [PMID: 39702476 DOI: 10.1038/s41537-024-00535-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/19/2024] [Indexed: 12/21/2024]
Abstract
Few studies using Positron Emission Tomography with 18F-fluorodeoxyglucose (18F-FDG-PET) have examined the neurobiological basis of antipsychotic resistance in schizophrenia, primarily focusing on metabolic activity, with none investigating connectivity patterns. Here, we aimed to explore differential patterns of glucose metabolism between patients and controls (CTRL) through a graph theory-based approach and network comparison tests. PET scans with 18F-FDG were obtained by 70 subjects, 26 with treatment-resistant schizophrenia (TRS), 28 patients responsive to antipsychotics (nTRS), and 16 CTRL. Relative brain glucose metabolism maps were processed in the automated anatomical labeling (AAL)-Merged atlas template. Inter-subject connectivity matrices were derived using Gaussian Graphical Models and group networks were compared through permutation testing. A logistic model based on machine-learning was employed to estimate the association between the metabolic signals of brain regions and treatment resistance. To account for the potential influence of antipsychotic medication, we incorporated chlorpromazine equivalents as a covariate in the network analysis during partial correlation calculations. Additionally, the machine-learning analysis employed medication dose-stratified folds. Global reduced connectivity was detected in the nTRS (p-value = 0.008) and TRS groups (p-value = 0.001) compared to CTRL, with prominent alterations localized in the frontal lobe, Default Mode Network, and dorsal dopamine pathway. Disruptions in frontotemporal and striatal-cortical connectivity were detected in TRS but not nTRS patients. After adjusting for antipsychotic doses, alterations in the anterior cingulate, frontal and temporal gyri, hippocampus, and precuneus also emerged. The machine-learning approach demonstrated an accuracy ranging from 0.72 to 0.8 in detecting the TRS condition.
Collapse
Affiliation(s)
- Giuseppe De Simone
- Section of Psychiatry, Laboratory of Molecular and Translational Psychiatry, Unit of Treatment-Resistant Psychiatric Disorders, Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples "Federico II", School of Medicine, Naples Italy, Via Pansini 5, 80131, Naples, Italy
| | - Felice Iasevoli
- Section of Psychiatry, Laboratory of Molecular and Translational Psychiatry, Unit of Treatment-Resistant Psychiatric Disorders, Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples "Federico II", School of Medicine, Naples Italy, Via Pansini 5, 80131, Naples, Italy
| | - Annarita Barone
- Section of Psychiatry, Laboratory of Molecular and Translational Psychiatry, Unit of Treatment-Resistant Psychiatric Disorders, Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples "Federico II", School of Medicine, Naples Italy, Via Pansini 5, 80131, Naples, Italy
| | - Valeria Gaudieri
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Via S. Pansini 5, 80131, Naples, Italy
| | - Alberto Cuocolo
- Department of Advanced Biomedical Sciences, University of Naples Federico II, Via S. Pansini 5, 80131, Naples, Italy
| | - Mariateresa Ciccarelli
- Section of Psychiatry, Laboratory of Molecular and Translational Psychiatry, Unit of Treatment-Resistant Psychiatric Disorders, Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples "Federico II", School of Medicine, Naples Italy, Via Pansini 5, 80131, Naples, Italy
| | - Sabina Pappatà
- Institute of Biostructure and Bioimaging, National Research Council, Via T. De Amicis 95, 80145, Naples, Italy
| | - Andrea de Bartolomeis
- Section of Psychiatry, Laboratory of Molecular and Translational Psychiatry, Unit of Treatment-Resistant Psychiatric Disorders, Department of Neuroscience, Reproductive Sciences and Dentistry, University of Naples "Federico II", School of Medicine, Naples Italy, Via Pansini 5, 80131, Naples, Italy.
| |
Collapse
|
2
|
Zahacy R, Ma Y, Winship IR, Jackson J, Chan AW. Claustrum modulation drives altered prefrontal cortex dynamics and connectivity. Commun Biol 2024; 7:1556. [PMID: 39578634 PMCID: PMC11584859 DOI: 10.1038/s42003-024-07256-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024] Open
Abstract
This study delves into the claustrum's role in modulating spontaneous and sensory-evoked network activity across cortical regions. Using mesoscale calcium imaging and Gi and Gq DREADDs in anesthetized mice, we show that decreasing claustral activity enhances prefrontal cortical activity, while activation reduces prefrontal cortical activity. This claustrum modulation also caused changes to the brain's large-scale functional networks, emphasizing the claustrum's ability to influence long-range functional connectivity in the cortex. Claustrum inhibition increased the local coupling between frontal cortex areas, but reduced the correlation between anterior medial regions and lateral/posterior regions, while also enhancing sensory-evoked responses in the visual cortex. These findings indicate the claustrum can participate in orchestrating neural communication across cortical regions through modulation of prefrontal cortical activity. These insights deepen our understanding of the claustrum's impact on prefrontal connectivity, large-scale network dynamics, and sensory processing, positioning the claustrum as a key node modulating large-scale cortical dynamics.
Collapse
Affiliation(s)
- Ryan Zahacy
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Yonglie Ma
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
| | - Ian R Winship
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada
| | - Jesse Jackson
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada.
- Department of Physiology, University of Alberta, Edmonton, AB, Canada.
| | - Allen W Chan
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada.
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
3
|
Hanania JU, Reimers E, Bevington CWJ, Sossi V. PET-based brain molecular connectivity in neurodegenerative disease. Curr Opin Neurol 2024; 37:353-360. [PMID: 38813843 DOI: 10.1097/wco.0000000000001283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
PURPOSE OF REVIEW Molecular imaging has traditionally been used and interpreted primarily in the context of localized and relatively static neurochemical processes. New understanding of brain function and development of novel molecular imaging protocols and analysis methods highlights the relevance of molecular networks that co-exist and interact with functional and structural networks. Although the concept and evidence of disease-specific metabolic brain patterns has existed for some time, only recently has such an approach been applied in the neurotransmitter domain and in the context of multitracer and multimodal studies. This review briefly summarizes initial findings and highlights emerging applications enabled by this new approach. RECENT FINDINGS Connectivity based approaches applied to molecular and multimodal imaging have uncovered molecular networks with neurodegeneration-related alterations to metabolism and neurotransmission that uniquely relate to clinical findings; better disease stratification paradigms; an improved understanding of the relationships between neurochemical and functional networks and their related alterations, although the directionality of these relationships are still unresolved; and a new understanding of the molecular underpinning of disease-related alteration in resting-state brain activity. SUMMARY Connectivity approaches are poised to greatly enhance the information that can be extracted from molecular imaging. While currently mostly contributing to enhancing understanding of brain function, they are highly likely to contribute to the identification of specific biomarkers that will improve disease management and clinical care.
Collapse
Affiliation(s)
| | - Erik Reimers
- Department of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | |
Collapse
|
4
|
Pedersen R, Johansson J, Nordin K, Rieckmann A, Wåhlin A, Nyberg L, Bäckman L, Salami A. Dopamine D1-Receptor Organization Contributes to Functional Brain Architecture. J Neurosci 2024; 44:e0621232024. [PMID: 38302439 PMCID: PMC10941071 DOI: 10.1523/jneurosci.0621-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 12/01/2023] [Accepted: 01/21/2024] [Indexed: 02/03/2024] Open
Abstract
Recent work has recognized a gradient-like organization in cortical function, spanning from primary sensory to transmodal cortices. It has been suggested that this axis is aligned with regional differences in neurotransmitter expression. Given the abundance of dopamine D1-receptors (D1DR), and its importance for modulation and neural gain, we tested the hypothesis that D1DR organization is aligned with functional architecture, and that inter-regional relationships in D1DR co-expression modulate functional cross talk. Using the world's largest dopamine D1DR-PET and MRI database (N = 180%, 50% female), we demonstrate that D1DR organization follows a unimodal-transmodal hierarchy, expressing a high spatial correspondence to the principal gradient of functional connectivity. We also demonstrate that individual differences in D1DR density between unimodal and transmodal regions are associated with functional differentiation of the apices in the cortical hierarchy. Finally, we show that spatial co-expression of D1DR primarily modulates couplings within, but not between, functional networks. Together, our results show that D1DR co-expression provides a biomolecular layer to the functional organization of the brain.
Collapse
Affiliation(s)
- Robin Pedersen
- Department of Integrative Medical Biology, Umeå University, Umeå S-90197, Sweden
- Wallenberg Center for Molecular Medicine (WCMM), Umeå University, Umeå S-90197, Sweden
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå S-90197, Sweden
| | - Jarkko Johansson
- Department of Integrative Medical Biology, Umeå University, Umeå S-90197, Sweden
- Wallenberg Center for Molecular Medicine (WCMM), Umeå University, Umeå S-90197, Sweden
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå S-90197, Sweden
| | - Kristin Nordin
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå S-90197, Sweden
- Aging Research Center, Karolinska Institutet & Stockholm University, Stockholm S-17165, Sweden
| | - Anna Rieckmann
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå S-90197, Sweden
- Department of Radiation Sciences, Umeå University, Umeå S-90197, Sweden
- Max-Planck-Institut für Sozialrecht und Sozialpolitik, Munich 80799, Germany
| | - Anders Wåhlin
- Department of Integrative Medical Biology, Umeå University, Umeå S-90197, Sweden
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå S-90197, Sweden
| | - Lars Nyberg
- Department of Integrative Medical Biology, Umeå University, Umeå S-90197, Sweden
- Wallenberg Center for Molecular Medicine (WCMM), Umeå University, Umeå S-90197, Sweden
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå S-90197, Sweden
- Department of Radiation Sciences, Umeå University, Umeå S-90197, Sweden
| | - Lars Bäckman
- Aging Research Center, Karolinska Institutet & Stockholm University, Stockholm S-17165, Sweden
| | - Alireza Salami
- Department of Integrative Medical Biology, Umeå University, Umeå S-90197, Sweden
- Wallenberg Center for Molecular Medicine (WCMM), Umeå University, Umeå S-90197, Sweden
- Umeå Center for Functional Brain Imaging (UFBI), Umeå University, Umeå S-90197, Sweden
- Aging Research Center, Karolinska Institutet & Stockholm University, Stockholm S-17165, Sweden
| |
Collapse
|
5
|
Chumin EJ, Burton CP, Silvola R, Miner EW, Persohn SC, Veronese M, Territo PR. Brain metabolic network covariance and aging in a mouse model of Alzheimer's disease. Alzheimers Dement 2024; 20:1538-1549. [PMID: 38032015 PMCID: PMC10984484 DOI: 10.1002/alz.13538] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 10/09/2023] [Accepted: 10/11/2023] [Indexed: 12/01/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD), the leading cause of dementia worldwide, represents a human and financial impact for which few effective drugs exist to treat the disease. Advances in molecular imaging have enabled assessment of cerebral glycolytic metabolism, and network modeling of brain region have linked to alterations in metabolic activity to AD stage. METHODS We performed 18 F-FDG positron emission tomography (PET) imaging in 4-, 6-, and 12-month-old 5XFAD and littermate controls (WT) of both sexes and analyzed region data via brain metabolic covariance analysis. RESULTS The 5XFAD model mice showed age-related changes in glucose uptake relative to WT mice. Analysis of community structure of covariance networks was different across age and sex, with a disruption of metabolic coupling in the 5XFAD model. DISCUSSION The current study replicates clinical AD findings and indicates that metabolic network covariance modeling provides a translational tool to assess disease progression in AD models.
Collapse
Affiliation(s)
- Evgeny J. Chumin
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of Psychological and Brain SciencesIndiana UniversityBloomingtonIndianaUSA
- Indiana University Network Science Institute, Indiana UniversityBloomingtonIndianaUSA
| | - Charles P. Burton
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Rebecca Silvola
- Department of MedicineDivision of Clinical PharmacologyIndiana University School of MedicineIndianapolisIndianaUSA
- Eli Lilly and CompanyIndianapolisIndianaUSA
| | - Ethan W. Miner
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Scott C. Persohn
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
| | - Mattia Veronese
- Department of Information EngineeringUniversity of PaduaPaduaItaly
- Department of NeuroimagingKing's College LondonLondonUK
| | - Paul R. Territo
- Stark Neurosciences Research InstituteIndiana University School of MedicineIndianapolisIndianaUSA
- Department of MedicineDivision of Clinical PharmacologyIndiana University School of MedicineIndianapolisIndianaUSA
| |
Collapse
|
6
|
Lizarraga A, Ripp I, Sala A, Shi K, Düring M, Koch K, Yakushev I. Similarity between structural and proxy estimates of brain connectivity. J Cereb Blood Flow Metab 2024; 44:284-295. [PMID: 37773727 PMCID: PMC10993877 DOI: 10.1177/0271678x231204769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 08/01/2023] [Accepted: 08/18/2023] [Indexed: 10/01/2023]
Abstract
Functional magnetic resonance and diffusion weighted imaging have so far made a major contribution to delineation of the brain connectome at the macroscale. While functional connectivity (FC) was shown to be related to structural connectivity (SC) to a certain degree, their spatial overlap is unknown. Even less clear are relations of SC with estimates of connectivity from inter-subject covariance of regional F18-fluorodeoxyglucose uptake (FDGcov) and grey matter volume (GMVcov). Here, we asked to what extent SC underlies three proxy estimates of brain connectivity: FC, FDGcov and GMVcov. Simultaneous PET/MR acquisitions were performed in 56 healthy middle-aged individuals. Similarity between four networks was assessed using Spearman correlation and convergence ratio (CR), a measure of spatial overlap. Spearman correlation coefficient was 0.27 for SC-FC, 0.40 for SC-FDGcov, and 0.15 for SC-GMVcov. Mean CRs were 51% for SC-FC, 48% for SC-FDGcov, and 37% for SC-GMVcov. These results proved to be reproducible and robust against image processing steps. In sum, we found a relevant similarity of SC with FC and FDGcov, while GMVcov consistently showed the weakest similarity. These findings indicate that white matter tracts underlie FDGcov to a similar degree as FC, supporting FDGcov as estimate of functional brain connectivity.
Collapse
Affiliation(s)
- Aldana Lizarraga
- Department of Nuclear Medicine, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Isabelle Ripp
- Department of Nuclear Medicine, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Arianna Sala
- Department of Nuclear Medicine, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
- Coma Science Group, GIGA Consciousness, University of Liege; Centre du Cerveau2, University Hospital of Liege, Avenue de L'Hôpital 1, Liege, Belgium
| | - Kuangyu Shi
- Department of Nuclear Medicine, University Hospital Bern, Bern, Switzerland
| | - Marco Düring
- Medical Image Analysis Center (MIAC AG) and Qbig, Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Kathrin Koch
- Department of Neuroradiology, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Igor Yakushev
- Department of Nuclear Medicine, School of Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
7
|
Burton CP, Chumin EJ, Collins AY, Persohn SA, Onos KD, Pandey RS, Quinney SK, Territo PR. Levetiracetam modulates brain metabolic networks and transcriptomic signatures in the 5XFAD mouse model of Alzheimer's disease. Front Neurosci 2024; 17:1336026. [PMID: 38328556 PMCID: PMC10847229 DOI: 10.3389/fnins.2023.1336026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 12/13/2023] [Indexed: 02/09/2024] Open
Abstract
Introduction Subcritical epileptiform activity is associated with impaired cognitive function and is commonly seen in patients with Alzheimer's disease (AD). The anti-convulsant, levetiracetam (LEV), is currently being evaluated in clinical trials for its ability to reduce epileptiform activity and improve cognitive function in AD. The purpose of the current study was to apply pharmacokinetics (PK), network analysis of medical imaging, gene transcriptomics, and PK/PD modeling to a cohort of amyloidogenic mice to establish how LEV restores or drives alterations in the brain networks of mice in a dose-dependent basis using the rigorous preclinical pipeline of the MODEL-AD Preclinical Testing Core. Methods Chronic LEV was administered to 5XFAD mice of both sexes for 3 months based on allometrically scaled clinical dose levels from PK models. Data collection and analysis consisted of a multi-modal approach utilizing 18F-FDG PET/MRI imaging and analysis, transcriptomic analyses, and PK/PD modeling. Results Pharmacokinetics of LEV showed a sex and dose dependence in Cmax, CL/F, and AUC0-∞, with simulations used to estimate dose regimens. Chronic dosing at 10, 30, and 56 mg/kg, showed 18F-FDG specific regional differences in brain uptake, and in whole brain covariance measures such as clustering coefficient, degree, network density, and connection strength (i.e., positive and negative). In addition, transcriptomic analysis via nanoString showed dose-dependent changes in gene expression in pathways consistent 18F-FDG uptake and network changes, and PK/PD modeling showed a concentration dependence for key genes, but not for network covariance modeling. Discussion This study represents the first report detailing the relationships of metabolic covariance and transcriptomic network changes resulting from LEV administration in 5XFAD mice. Overall, our results highlight non-linear kinetics based on dose and sex, where gene expression analysis demonstrated LEV dose- and concentration-dependent changes, along with cerebral metabolism, and/or cerebral homeostatic mechanisms relevant to human AD, which aligned closely with network covariance analysis of 18F-FDG images. Collectively, this study show cases the value of a multimodal connectomic, transcriptomic, and pharmacokinetic approach to further investigate dose dependent relationships in preclinical studies, with translational value toward informing clinical study design.
Collapse
Affiliation(s)
- Charles P. Burton
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Evgeny J. Chumin
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Alyssa Y. Collins
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Scott A. Persohn
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| | | | - Ravi S. Pandey
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| | - Sara K. Quinney
- Department of Medicine, Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Paul R. Territo
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
- Department of Medicine, Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
8
|
Burton CP, Chumin EJ, Collins AY, Persohn SA, Onos KD, Pandey RS, Quinney SK, Territo PR. Levetiracetam Modulates Brain Metabolic Networks and Transcriptomic Signatures in the 5XFAD Mouse Model of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.10.566574. [PMID: 38014102 PMCID: PMC10680636 DOI: 10.1101/2023.11.10.566574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
INTRODUCTION Subcritical epileptiform activity is associated with impaired cognitive function and is commonly seen in patients with Alzheimer's disease (AD). The anti-convulsant, levetiracetam (LEV), is currently being evaluated in clinical trials for its ability to reduce epileptiform activity and improve cognitive function in AD. The purpose of the current study was to apply pharmacokinetics (PK), network analysis of medical imaging, gene transcriptomics, and PK/PD modeling to a cohort of amyloidogenic mice to establish how LEV restores or drives alterations in the brain networks of mice in a dose-dependent basis using the rigorous preclinical pipeline of the MODEL-AD Preclinical Testing Core. METHODS Chronic LEV was administered to 5XFAD mice of both sexes for 3 months based on allometrically scaled clinical dose levels from PK models. Data collection and analysis consisted of a multi-modal approach utilizing 18F-FDG PET/MRI imaging and analysis, transcriptomic analyses, and PK/PD modeling. RESULTS Pharmacokinetics of LEV showed a sex and dose dependence in Cmax, CL/F, and AUC0-∞, with simulations used to estimate dose regimens. Chronic dosing at 10, 30, and 56 mg/kg, showed 18F-FDG specific regional differences in brain uptake, and in whole brain covariance measures such as clustering coefficient, degree, network density, and connection strength (i.e. positive and negative). In addition, transcriptomic analysis via nanoString showed dose-dependent changes in gene expression in pathways consistent 18F-FDG uptake and network changes, and PK/PD modeling showed a concentration dependence for key genes, but not for network covariance modeling. DISCUSSION This study represents the first report detailing the relationships of metabolic covariance and transcriptomic network changes resulting from LEV administration in 5XFAD mice. Overall, our results highlight non-linear kinetics based on dose and sex, where gene expression analysis demonstrated LEV dose- and concentration- dependent changes, along with cerebral metabolism, and/or cerebral homeostatic mechanisms relevant to human AD, which aligned closely with network covariance analysis of 18F-FDG images. Collectively, this study show cases the value of a multimodal connectomic, transcriptomic, and pharmacokinetic approach to further investigate dose dependent relationships in preclinical studies, with translational value towards informing clinical study design.
Collapse
Affiliation(s)
- Charles P. Burton
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis IN 46202 USA
| | - Evgeny J. Chumin
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis IN 46202 USA
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis IN 46202
| | - Alyssa Y. Collins
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis IN 46202 USA
| | - Scott A. Persohn
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis IN 46202 USA
| | | | - Ravi S. Pandey
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06032
| | - Sara K. Quinney
- Department of Medicine, Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis IN 46202 USA
| | - Paul R. Territo
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis IN 46202 USA
- Department of Medicine, Division of Clinical Pharmacology, Indiana University School of Medicine, Indianapolis IN 46202 USA
| |
Collapse
|
9
|
Volpi T, Vallini G, Silvestri E, Francisci MD, Durbin T, Corbetta M, Lee JJ, Vlassenko AG, Goyal MS, Bertoldo A. A new framework for metabolic connectivity mapping using bolus [ 18F]FDG PET and kinetic modeling. J Cereb Blood Flow Metab 2023; 43:1905-1918. [PMID: 37377103 PMCID: PMC10676136 DOI: 10.1177/0271678x231184365] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 04/11/2023] [Accepted: 06/03/2023] [Indexed: 06/29/2023]
Abstract
Metabolic connectivity (MC) has been previously proposed as the covariation of static [18F]FDG PET images across participants, i.e., across-individual MC (ai-MC). In few cases, MC has been inferred from dynamic [18F]FDG signals, i.e., within-individual MC (wi-MC), as for resting-state fMRI functional connectivity (FC). The validity and interpretability of both approaches is an important open issue. Here we reassess this topic, aiming to 1) develop a novel wi-MC methodology; 2) compare ai-MC maps from standardized uptake value ratio (SUVR) vs. [18F]FDG kinetic parameters fully describing the tracer behavior (i.e., Ki, K1, k3); 3) assess MC interpretability in comparison to structural connectivity and FC. We developed a new approach based on Euclidean distance to calculate wi-MC from PET time-activity curves. The across-individual correlation of SUVR, Ki, K1, k3 produced different networks depending on the chosen [18F]FDG parameter (k3 MC vs. SUVR MC, r = 0.44). We found that wi-MC and ai-MC matrices are dissimilar (maximum r = 0.37), and that the match with FC is higher for wi-MC (Dice similarity: 0.47-0.63) than for ai-MC (0.24-0.39). Our analyses demonstrate that calculating individual-level MC from dynamic PET is feasible and yields interpretable matrices that bear similarity to fMRI FC measures.
Collapse
Affiliation(s)
- Tommaso Volpi
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
- Padova Neuroscience Center, University of Padova, Padova, Italy
| | - Giulia Vallini
- Department of Information Engineering, University of Padova, Padova, Italy
| | - Erica Silvestri
- Department of Information Engineering, University of Padova, Padova, Italy
| | | | - Tony Durbin
- Neuroimaging Laboratories at the Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Maurizio Corbetta
- Padova Neuroscience Center, University of Padova, Padova, Italy
- Department of Neuroscience, University of Padova, Padova, Italy
| | - John J Lee
- Neuroimaging Laboratories at the Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Andrei G Vlassenko
- Neuroimaging Laboratories at the Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Manu S Goyal
- Neuroimaging Laboratories at the Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, MO, USA
| | - Alessandra Bertoldo
- Padova Neuroscience Center, University of Padova, Padova, Italy
- Department of Information Engineering, University of Padova, Padova, Italy
| |
Collapse
|
10
|
Carli G, Cavicchioli M, Martini AL, Bruscoli M, Manfredi A, Presotto L, Mazzeo C, Sestini S, Perani D. Neurobiological Dysfunctional Substrates for the Self-Medication Hypothesis in Adult Individuals with Attention-Deficit Hyperactivity Disorder and Cocaine Use Disorder: A Fluorine-18-Fluorodeoxyglucose Positron Emission Tomography Study. Brain Connect 2023; 13:370-382. [PMID: 37097207 DOI: 10.1089/brain.2022.0076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023] Open
Abstract
Objectives: Attention-deficit hyperactivity disorder (ADHD) in adulthood shows high co-occurrence rates with cocaine use disorder (CoUD). The self-medication hypothesis (SMH) provides a theoretical explanation for this comorbidity. This study investigates the neurobiological mechanisms that could support SMH in adult patients with attention-deficit hyperactivity disorder with cocaine use disorder (ADHD-CoUD). Materials and Methods: We included 19 ADHD-CoUD patients (84.2% male; age: 32.11 years [7.18]) and 16 CoUD patients (68.7% male; age: 36.63 years [8.12]). All subjects underwent a fluorine-18-fluorodeoxyglucose positron emission tomography (18F-FDG PET) brain scan. We tested brain metabolism differences between ADHD-CoUD and CoUD patients using voxel-based and regions of interest (ROIs)-based analyses. The correlation between dependence/abstinence duration and regional brain metabolism was also assessed in the two groups. Lastly, we investigated the integrity of brain metabolic connectivity of mesocorticolimbic and nigrostriatal dopaminergic systems, and large-scale brain networks involved in ADHD and addictions. Results: The voxel-wise and ROIs-based approaches showed that ADHD-CoUD patients had a lower metabolism in the thalamus and increased metabolism in the amygdala and parahippocampus, bilaterally, than CoUD subjects and healthy controls (HCs). Metabolism in the thalamus negatively correlated with years of dependence in ADHD-CoUD patients. Moreover, connectivity analyses revealed that ADHD-CoUD patients had a more preserved metabolic connectivity than CoUD patients in the dopaminergic networks and large-scale networks involved in self-regulation mechanisms of attention and behaviors (i.e., anterior default mode network [ADMN], executive network [ECN], and anterior salience network [aSAN]). Conclusions: We demonstrated distinct neuropathological substrates underlying substance-use behaviors in ADHD-CoUD and CoUD patients. Furthermore, we provided neurobiological evidence in support of SMH, demonstrating that ADHD-CoUD patients might experience short-term advantages of cocaine assumption (i.e., compensation of dopaminergic deficiency and related cognitive-behavioral deficits).
Collapse
Affiliation(s)
- Giulia Carli
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marco Cavicchioli
- Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy
| | - Anna Lisa Martini
- Unit of Nuclear Medicine, Department of Diagnostic Imaging, N.O.P.-S. Stefano, U.S.L. Toscana Centro, Prato, Italy
| | - Matteo Bruscoli
- UFC Farmacotossicodipendenze, Department of Drug Addiction, N.O.P.-S. Stefano, U.S.L. Toscana Centro, Prato, Italy
| | - Antonella Manfredi
- UFC Farmacotossicodipendenze, Department of Drug Addiction, N.O.P.-S. Stefano, U.S.L. Toscana Centro, Prato, Italy
| | - Luca Presotto
- Department of Physics G. Occhialini, Università degli Studi di Milano Bicocca, Milan, Italy
| | - Christian Mazzeo
- Unit of Nuclear Medicine, Department of Diagnostic Imaging, N.O.P.-S. Stefano, U.S.L. Toscana Centro, Prato, Italy
| | - Stelvio Sestini
- Unit of Nuclear Medicine, Department of Diagnostic Imaging, N.O.P.-S. Stefano, U.S.L. Toscana Centro, Prato, Italy
| | - Daniela Perani
- Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy
- In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Nuclear Medicine Unit, San Raffaele Hospital, Milan, Italy
| |
Collapse
|
11
|
Drake DF, Derado G, Zhang L, Bowman FD. Neuroimaging statistical approaches for determining neural correlates of Alzheimer's disease via positron emission tomography imaging. WILEY INTERDISCIPLINARY REVIEWS. COMPUTATIONAL STATISTICS 2023; 15:e1606. [PMID: 39655245 PMCID: PMC11626230 DOI: 10.1002/wics.1606] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 01/05/2023] [Indexed: 12/12/2024]
Abstract
Alzheimer's disease (AD) is a degenerative disorder involving significant memory loss and other cognitive deficits, manifesting as a progression from normal cognitive functioning to mild cognitive impairment to AD. The sooner an accurate diagnosis of probable AD is made, the easier it is to manage symptoms and plan for future therapy. Functional neuroimaging stands to be a useful tool in achieving early diagnosis. Among the many neuroimaging modalities, positron emission tomography (PET) provides direct regional assessment of, among others, brain metabolism, cerebral blood flow, amyloid deposition-all quantities of interest in the characterization of AD. However, there are analytic challenges in identifying early indicators of AD from these high-dimensional imaging data sets, and it is unclear whether early indicators of AD are more likely to emerge in localized patterns of brain activity or in patterns of correlation between distinct brain regions. Early PET-based analyses of AD focused on alterations in metabolic activity at the voxel-level or in anatomically defined regions of interest. Other approaches, including seed-voxel and multivariate techniques, seek to characterize metabolic connectivity by identifying other regions in the brain with similar patterns of activity across subjects. We briefly review various neuroimaging statistical approaches applied to determine changes in metabolic activity or metabolic connectivity associated with AD. We then present an approach that provides a unified statistical framework for addressing both metabolic activity and connectivity. Specifically, we apply a Bayesian spatial hierarchical framework to longitudinal metabolic PET scans from the Alzheimer's Disease Neuroimaging Initiative.
Collapse
Affiliation(s)
- Daniel F. Drake
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Gordana Derado
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Lijun Zhang
- Department of Population and Quantitative Health Science, Case Western Reserve University, Cleveland, Ohio, USA
| | - F. DuBois Bowman
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | | |
Collapse
|
12
|
Sala A, Lizarraga A, Caminiti SP, Calhoun VD, Eickhoff SB, Habeck C, Jamadar SD, Perani D, Pereira JB, Veronese M, Yakushev I. Brain connectomics: time for a molecular imaging perspective? Trends Cogn Sci 2023; 27:353-366. [PMID: 36621368 PMCID: PMC10432882 DOI: 10.1016/j.tics.2022.11.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/19/2022] [Accepted: 11/30/2022] [Indexed: 01/09/2023]
Abstract
In the past two decades brain connectomics has evolved into a major concept in neuroscience. However, the current perspective on brain connectivity and how it underpins brain function relies mainly on the hemodynamic signal of functional magnetic resonance imaging (MRI). Molecular imaging provides unique information inaccessible to MRI-based and electrophysiological techniques. Thus, positron emission tomography (PET) has been successfully applied to measure neural activity, neurotransmission, and proteinopathies in normal and pathological cognition. Here, we position molecular imaging within the brain connectivity framework from the perspective of timeliness, validity, reproducibility, and resolution. We encourage the neuroscientific community to take an integrative approach whereby MRI-based, electrophysiological techniques, and molecular imaging contribute to our understanding of the brain connectome.
Collapse
Affiliation(s)
- Arianna Sala
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technical University of Munich, School of Medicine, 81675 Munich, Germany; Coma Science Group, GIGA-Consciousness, University of Liege, 4000 Liege, Belgium; Centre du Cerveau(2), University Hospital of Liege, 4000 Liege, Belgium
| | - Aldana Lizarraga
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technical University of Munich, School of Medicine, 81675 Munich, Germany
| | - Silvia Paola Caminiti
- Vita-Salute San Raffaele University, 20132 Milan, Italy; In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Vince D Calhoun
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia State University, Georgia Institute of Technology, and Emory University, Atlanta, GA 30303, USA
| | - Simon B Eickhoff
- Institute of Neuroscience and Medicine, Brain, and Behaviour (INM-7), Research Centre Jülich, 52428 Jülich, Germany; Institute of Systems Neuroscience, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany
| | - Christian Habeck
- Cognitive Neuroscience Division, Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Sharna D Jamadar
- Turner Institute for Brain and Mental Health, Monash University, 3800 Melbourne, Australia; Monash Biomedical Imaging, Monash University, 3800 Melbourne, Australia
| | - Daniela Perani
- Vita-Salute San Raffaele University, 20132 Milan, Italy; In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Raffaele Scientific Institute, 20132 Milan, Italy; Nuclear Medicine Unit, San Raffaele Hospital, 20132 Milan, Italy
| | - Joana B Pereira
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 14152 Stockholm, Sweden; Memory Research Unit, Department of Clinical Sciences, Malmö Lund University, 20502 Lund, Sweden
| | - Mattia Veronese
- Department of Neuroimaging, King's College London, London SE5 8AF, UK; Department of Information Engineering, University of Padua, 35131 Padua, Italy
| | - Igor Yakushev
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technical University of Munich, School of Medicine, 81675 Munich, Germany.
| |
Collapse
|
13
|
Protas H, Ghisays V, Goradia DD, Bauer R, Devadas V, Chen K, Reiman EM, Su Y. Individualized network analysis: A novel approach to investigate tau PET using graph theory in the Alzheimer's disease continuum. Front Neurosci 2023; 17:1089134. [PMID: 36937677 PMCID: PMC10017746 DOI: 10.3389/fnins.2023.1089134] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 02/14/2023] [Indexed: 03/06/2023] Open
Abstract
Introduction Tau PET imaging has emerged as an important tool to detect and monitor tangle burden in vivo in the study of Alzheimer's disease (AD). Previous studies demonstrated the association of tau burden with cognitive decline in probable AD cohorts. This study introduces a novel approach to analyze tau PET data by constructing individualized tau network structure and deriving its graph theory-based measures. We hypothesize that the network- based measures are a measure of the total tau load and the stage through disease. Methods Using tau PET data from the AD Neuroimaging Initiative from 369 participants, we determine the network measures, global efficiency, global strength, and limbic strength, and compare with two regional measures entorhinal and tau composite SUVR, in the ability to differentiate, cognitively unimpaired (CU), MCI and AD. We also investigate the correlation of these network and regional measures and a measure of memory performance, auditory verbal learning test for long-term recall memory (AVLT-LTM). Finally, we determine the stages based on global efficiency and limbic strength using conditional inference trees and compare with Braak staging. Results We demonstrate that the derived network measures are able to differentiate three clinical stages of AD, CU, MCI, and AD. We also demonstrate that these network measures are strongly correlated with memory performance overall. Unlike regional tau measurements, the tau network measures were significantly associated with AVLT-LTM even in cognitively unimpaired individuals. Stages determined from global efficiency and limbic strength, visually resembled Braak staging. Discussion The strong correlations with memory particularly in CU suggest the proposed technique may be used to characterize subtle early tau accumulation. Further investigation is ongoing to examine this technique in a longitudinal setting.
Collapse
Affiliation(s)
- Hillary Protas
- Banner Alzheimer’s Institute, Phoenix, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Valentina Ghisays
- Banner Alzheimer’s Institute, Phoenix, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Dhruman D. Goradia
- Banner Alzheimer’s Institute, Phoenix, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Robert Bauer
- Banner Alzheimer’s Institute, Phoenix, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Vivek Devadas
- Banner Alzheimer’s Institute, Phoenix, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
| | - Kewei Chen
- Banner Alzheimer’s Institute, Phoenix, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
- Department of Neurology, The University of Arizona, Tucson, AZ, United States
- Department of Psychiatry, The University of Arizona, Tucson, AZ, United States
- Department of Neuroscience, School of Computing and Augmented Intelligence, Biostatistical Core, School of Mathematics and Statistics, College of Health Solutions, Arizona State University, Tempe, AZ, United States
| | - Eric M. Reiman
- Banner Alzheimer’s Institute, Phoenix, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
- Department of Neurology, The University of Arizona, Tucson, AZ, United States
- Department of Psychiatry, The University of Arizona, Tucson, AZ, United States
- Department of Neuroscience, School of Computing and Augmented Intelligence, Biostatistical Core, School of Mathematics and Statistics, College of Health Solutions, Arizona State University, Tempe, AZ, United States
- Translational Genomics Research Institute, Phoenix, AZ, United States
| | - Yi Su
- Banner Alzheimer’s Institute, Phoenix, AZ, United States
- Arizona Alzheimer’s Consortium, Phoenix, AZ, United States
- Department of Neuroscience, School of Computing and Augmented Intelligence, Biostatistical Core, School of Mathematics and Statistics, College of Health Solutions, Arizona State University, Tempe, AZ, United States
| |
Collapse
|
14
|
Tassan Mazzocco M, Murtaj V, Martins D, Schellino R, Coliva A, Toninelli E, Vercelli A, Turkheimer F, Belloli S, Moresco RM. Exploring the neuroprotective effects of montelukast on brain inflammation and metabolism in a rat model of quinolinic acid-induced striatal neurotoxicity. J Neuroinflammation 2023; 20:34. [PMID: 36782185 PMCID: PMC9923670 DOI: 10.1186/s12974-023-02714-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 01/31/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND One intrastriatal administration of quinolinic acid (QA) in rats induces a lesion with features resembling those observed in Huntington's disease. Our aim is to evaluate the effects of the cysteinyl leukotriene receptor antagonist montelukast (MLK), which exhibited neuroprotection in different preclinical models of neurodegeneration, on QA-induced neuroinflammation and regional metabolic functions. METHODS The right and left striatum of Sprague Dawley and athymic nude rats were injected with QA and vehicle (VEH), respectively. Starting from the day before QA injection, animals were treated with 1 or 10 mg/kg of MLK or VEH for 14 days. At 14 and 30 days post-lesion, animals were monitored with magnetic resonance imaging (MRI) and positron emission tomography (PET) using [18F]-VC701, a translocator protein (TSPO)-specific radiotracer. Striatal neuroinflammatory response was measured post-mortem in rats treated with 1 mg/kg of MLK by immunofluorescence. Rats treated with 10 mg/kg of MLK also underwent a [18F]-FDG PET study at baseline and 4 months after lesion. [18F]-FDG PET data were then used to assess metabolic connectivity between brain regions by applying a covariance analysis method. RESULTS MLK treatment was not able to reduce the QA-induced increase in striatal TSPO PET signal and MRI lesion volume, where we only detected a trend towards reduction in animals treated with 10 mg/kg of MLK. Post-mortem immunofluorescence analysis revealed that MLK attenuated the increase in striatal markers of astrogliosis and activated microglia in the lesioned hemisphere. We also found a significant increase in a marker of anti-inflammatory activity (MannR) and a trend towards reduction in a marker of pro-inflammatory activity (iNOS) in the lesioned striatum of MLK-compared to VEH-treated rats. [18F]-FDG uptake was significantly reduced in the striatum and ipsilesional cortical regions of VEH-treated rats at 4 months after lesion. MLK administration preserved glucose metabolism in these cortical regions, but not in the striatum. Finally, MLK was able to counteract changes in metabolic connectivity and measures of network topology induced by QA, in both lesioned and non-lesioned hemispheres. CONCLUSIONS Overall, MLK treatment produced a significant neuroprotective effect by reducing neuroinflammation assessed by immunofluorescence and preserving regional brain metabolism and metabolic connectivity from QA-induced neurotoxicity in cortical and subcortical regions.
Collapse
Affiliation(s)
- Margherita Tassan Mazzocco
- PhD Program in Neuroscience, Medicine and Surgery Department, University of Milano-Bicocca, Milan, Italy
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Valentina Murtaj
- PhD Program in Neuroscience, Medicine and Surgery Department, University of Milano-Bicocca, Milan, Italy
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Daniel Martins
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Roberta Schellino
- Department of Neuroscience "Rita Levi Montalcini" and Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Turin, Italy
| | - Angela Coliva
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Elisa Toninelli
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Alessandro Vercelli
- Department of Neuroscience "Rita Levi Montalcini" and Neuroscience Institute Cavalieri Ottolenghi, University of Turin, Turin, Italy
| | - Federico Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Sara Belloli
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy
| | - Rosa Maria Moresco
- Nuclear Medicine Department, San Raffaele Scientific Institute (IRCCS), Milan, Italy.
- Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Milan, Italy.
- Technomed Foundation and Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy.
| |
Collapse
|
15
|
Rauchmann B, Brendel M, Franzmeier N, Trappmann L, Zaganjori M, Ersoezlue E, Morenas‐Rodriguez E, Guersel S, Burow L, Kurz C, Haeckert J, Tatò M, Utecht J, Papazov B, Pogarell O, Janowitz D, Buerger K, Ewers M, Palleis C, Weidinger E, Biechele G, Schuster S, Finze A, Eckenweber F, Rupprecht R, Rominger A, Goldhardt O, Grimmer T, Keeser D, Stoecklein S, Dietrich O, Bartenstein P, Levin J, Höglinger G, Perneczky R. Microglial activation and connectivity in Alzheimer's disease and aging. Ann Neurol 2022; 92:768-781. [DOI: 10.1002/ana.26465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Boris‐Stephan Rauchmann
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
- Sheffield Institute for Translational Neuroscience (SITraN) University of Sheffield Sheffield UK
- Department of Neuroradiology University Hospital LMU Munich Germany
| | - Matthias Brendel
- Department of Nuclear Medicine University Hospital, LMU Munich Munich Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich Germany
| | - Nicolai Franzmeier
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich Munich Germany
| | - Lena Trappmann
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Mirlind Zaganjori
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Ersin Ersoezlue
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Estrella Morenas‐Rodriguez
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
- Chair of Metabolic Biochemistry, Biomedical Center (BMC), Faculty of Medicine, LMU Munich Munich Germany
| | - Selim Guersel
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
| | - Lena Burow
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Carolin Kurz
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Jan Haeckert
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
- Department of Psychiatry, Psychotherapy and Psychosomatics University of Augsburg, Bezirkskrankenhaus Augsburg Augsburg Germany
| | - Maia Tatò
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Julia Utecht
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Boris Papazov
- Department of Radiology University Hospital, LMU Munich Munich Germany
| | - Oliver Pogarell
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich Munich Germany
| | - Katharina Buerger
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich Munich Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
| | - Michael Ewers
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich Munich Germany
| | - Carla Palleis
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
- Department of Neurology University Hospital, LMU Munich Munich Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich Germany
| | - Endy Weidinger
- Department of Neurology University Hospital, LMU Munich Munich Germany
| | - Gloria Biechele
- Department of Nuclear Medicine University Hospital, LMU Munich Munich Germany
| | - Sebastian Schuster
- Department of Nuclear Medicine University Hospital, LMU Munich Munich Germany
| | - Anika Finze
- Department of Nuclear Medicine University Hospital, LMU Munich Munich Germany
| | - Florian Eckenweber
- Department of Nuclear Medicine University Hospital, LMU Munich Munich Germany
| | - Rainer Rupprecht
- Department of Psychiatry and Psychotherapy University of Regensburg Regensburg Germany
| | - Axel Rominger
- Department of Nuclear Medicine University Hospital, LMU Munich Munich Germany
- Department of Nuclear Medicine University of Bern, Inselspital Bern Switzerland
| | - Oliver Goldhardt
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar Technical University Munich Munich Germany
| | - Timo Grimmer
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar Technical University Munich Munich Germany
| | - Daniel Keeser
- Department of Radiology University Hospital, LMU Munich Munich Germany
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
- Department of Neuroradiology University Hospital LMU Munich Germany
| | - Sophia Stoecklein
- Department of Radiology University Hospital, LMU Munich Munich Germany
| | - Olaf Dietrich
- Department of Radiology University Hospital, LMU Munich Munich Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine University Hospital, LMU Munich Munich Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich Germany
| | - Johannes Levin
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
- Department of Neurology University Hospital, LMU Munich Munich Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich Germany
| | - Günter Höglinger
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
- Department of Neurology Hannover Medical School Hannover Germany
| | - Robert Perneczky
- Department of Psychiatry and Psychotherapy University Hospital, LMU Munich Munich Germany
- German Center for Neurodegenerative Diseases (DZNE) Munich Munich Germany
- Ageing Epidemiology (AGE) Research Unit, School of Public Health Imperial College London London UK
- Munich Cluster for Systems Neurology (SyNergy), Munich Germany
- Sheffield Institute for Translational Neuroscience (SITraN) University of Sheffield Sheffield UK
| |
Collapse
|
16
|
Sun T, Wang Z, Wu Y, Gu F, Li X, Bai Y, Shen C, Hu Z, Liang D, Liu X, Zheng H, Yang Y, El Fakhri G, Zhou Y, Wang M. Identifying the individual metabolic abnormities from a systemic perspective using whole-body PET imaging. Eur J Nucl Med Mol Imaging 2022; 49:2994-3004. [PMID: 35567627 PMCID: PMC9106794 DOI: 10.1007/s00259-022-05832-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 05/01/2022] [Indexed: 12/28/2022]
Abstract
Introduction Distinct physiological states arise from complex interactions among the various organs present in the human body. PET is a non-invasive modality with numerous successful applications in oncology, neurology, and cardiology. However, while PET imaging has been applied extensively in detecting focal lesions or diseases, its potential in detecting systemic abnormalities is seldom explored, mostly because total-body imaging was not possible until recently. Methods In this context, the present study proposes a framework capable of constructing an individual metabolic abnormality network using a subject’s whole-body 18F-FDG SUV image and a normal control database. The developed framework was evaluated in the patients with lung cancer, the one discharged after suffering from Covid-19 disease, and the one that had gastrointestinal bleeding with the underlying cause unknown. Results The framework could successfully capture the deviation of these patients from healthy subjects at the level of both system and organ. The strength of the altered network edges revealed the abnormal metabolic connection between organs. The overall deviation of the network nodes was observed to be highly correlated to the organ SUV measures. Therefore, the molecular connectivity of glucose metabolism was characterized at a single subject level. Conclusion The proposed framework represents a significant step toward the use of PET imaging for identifying metabolic dysfunction from a systemic perspective. A better understanding of the underlying biological mechanisms and the physiological interpretation of the interregional connections identified in the present study warrant further research.
Collapse
Affiliation(s)
- Tao Sun
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China.
| | - Zhenguo Wang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China
| | - Yaping Wu
- Department of Medical Imaging, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Fengyun Gu
- Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, People's Republic of China
- Department of Statistics, School of Mathematical Sciences, University College Cork, Cork, Ireland
| | - Xiaochen Li
- Department of Medical Imaging, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Yan Bai
- Department of Medical Imaging, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| | - Chushu Shen
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China
| | - Zhanli Hu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China
| | - Dong Liang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China
| | - Xin Liu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China
| | - Yongfeng Yang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, People's Republic of China
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Yun Zhou
- Central Research Institute, United Imaging Healthcare Group Co., Ltd, Shanghai, People's Republic of China
- School of Biomedical Engineering, Shanghai Tech University, Shanghai, People's Republic of China
| | - Meiyun Wang
- Department of Medical Imaging, Henan Provincial People's Hospital and People's Hospital of Zhengzhou University, Zhengzhou, People's Republic of China.
| |
Collapse
|
17
|
Martins D, Brodmann K, Veronese M, Dipasquale O, Mazibuko N, Schuschnig U, Zelaya F, Fotopoulou A, Paloyelis Y. "Less is more": a dose-response account of intranasal oxytocin pharmacodynamics in the human brain. Prog Neurobiol 2022; 211:102239. [PMID: 35122880 DOI: 10.1016/j.pneurobio.2022.102239] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/23/2022] [Accepted: 01/31/2022] [Indexed: 12/27/2022]
Abstract
Intranasal oxytocin is attracting attention as a potential treatment for several brain disorders due to promising preclinical results. However, translating findings to humans has been hampered by remaining uncertainties about its pharmacodynamics and the methods used to probe its effects in the human brain. Using a dose-response design (9, 18 and 36 IU), we demonstrate that intranasal oxytocin-induced changes in local regional cerebral blood flow (rCBF) in the amygdala at rest, and in the covariance between rCBF in the amygdala and other key hubs of the brain oxytocin system, follow a dose-response curve with maximal effects for lower doses. Yet, the effects on local rCBF might vary by amygdala subdivision, highlighting the need to qualify dose-response curves within subregion. We further link physiological changes with the density of the oxytocin receptor gene mRNA across brain regions, strengthening our confidence in intranasal oxytocin as a valid approach to engage central targets. Finally, we demonstrate that intranasal oxytocin does not disrupt cerebrovascular reactivity, which corroborates the validity of haemodynamic neuroimaging to probe the effects of intranasal oxytocin in the human brain. DATA AVAILABILITY: Participants did not consent for open sharing of the data. Therefore, data can only be accessed from the corresponding author upon reasonable request.
Collapse
Affiliation(s)
- Daniel Martins
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Katja Brodmann
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Mattia Veronese
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Ottavia Dipasquale
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Ndaba Mazibuko
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | | | - Fernando Zelaya
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK
| | - Aikaterini Fotopoulou
- Department of Clinical, Educational and Health Psychology, University College London, London, UK
| | - Yannis Paloyelis
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, De Crespigny Park, London SE5 8AF, UK.
| |
Collapse
|
18
|
NRM 2021 Abstract Booklet. J Cereb Blood Flow Metab 2021; 41:11-309. [PMID: 34905986 PMCID: PMC8851538 DOI: 10.1177/0271678x211061050] [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: 11/15/2022]
|
19
|
Volpi T, Silvestri E, Corbetta M, Bertoldo A. Assessing different approaches to estimate single-subject metabolic connectivity from dynamic [ 18F]fluorodeoxyglucose Positron Emission Tomography data. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2021; 2021:3259-3262. [PMID: 34891936 DOI: 10.1109/embc46164.2021.9630441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Metabolic connectivity is conventionally calculated in terms of correlation of static positron emission tomography (PET) measurements across subjects. There is increasing interest in deriving metabolic connectivity at the single-subject level from dynamic PET data, in a similar way to functional magnetic resonance imaging. However, the strong multicollinearity among region-wise PET time-activity curves (TACs), their non-Gaussian distribution, and the choice of the best strategy for TAC standardization before metabolic connectivity estimation, are non-trivial methodological issues to be tackled.In this work we test four different approaches to estimate sparse inverse covariance matrices, as well as three similarity-based methods to derive adjacency matrices. These approaches, combined with three different TAC standardization strategies, are employed to quantify metabolic connectivity from dynamic [18F]fluorodeoxyglucose ([18F]FDG) PET data in four healthy subjects.
Collapse
|
20
|
Sala A, Lizarraga A, Ripp I, Cumming P, Yakushev I. Static versus Functional PET: Making Sense of Metabolic Connectivity. Cereb Cortex 2021; 32:1125-1129. [PMID: 34411237 DOI: 10.1093/cercor/bhab271] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/16/2021] [Accepted: 07/16/2021] [Indexed: 11/13/2022] Open
Abstract
Recently, Jamadar et al. (2021, Metabolic and hemodynamic resting-state connectivity of the human brain: a high-temporal resolution simultaneous BOLD-fMRI and FDG-fPET multimodality study. Cereb Cortex. 31(6), 2855-2867) compared the patterns of brain connectivity or covariance as obtained from 3 neuroimaging measures: 1) functional connectivity estimated from temporal correlations in the functional magnetic resonance imaging blood oxygen level-dependent signal, metabolic connectivity estimated, 2) from temporal correlations in 16-s frames of dynamic [18F]-fluorodeoxyglucose-positron emission tomography (FDG-PET), which they designate as functional FDG-PET (fPET), and 3) from intersubject correlations in static FDG-PET images (sPET). Here, we discuss a number of fundamental issues raised by the Jamadar study. These include the choice of terminology, the interpretation of cross-modal findings, the issue of group- to single-subject level inferences, and the meaning of metabolic connectivity as a biomarker. We applaud the methodological approach taken by the authors, but wish to present an alternative perspective on their findings. In particular, we argue that sPET and fPET can both provide valuable information about brain connectivity. Certainly, resolving this conundrum calls for further experimental and theoretical efforts to advance the developing framework of PET-based brain connectivity indices.
Collapse
Affiliation(s)
- Arianna Sala
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich 81675, Germany.,Coma Science Group, GIGA Consciousness, University of Liege, Liege 4000, Belgium.,Centre du Cerveau2, University Hospital of Liege, Liege 4000, Belgium
| | - Aldana Lizarraga
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich 81675, Germany.,Neuroimaging Center (TUM-NIC), Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich 81675, Germany
| | - Isabelle Ripp
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich 81675, Germany.,Neuroimaging Center (TUM-NIC), Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich 81675, Germany.,Graduate School of Systemic Neurosciences, Ludwig-Maximilians-University, Planegg 82152, Germany
| | - Paul Cumming
- Department of Nuclear Medicine, Bern University Hospital, Bern 3010, Switzerland.,School of Psychology and Counselling, Queensland University of Technology, Brisbane 4059, Australia
| | - Igor Yakushev
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich 81675, Germany.,Neuroimaging Center (TUM-NIC), Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich 81675, Germany.,Graduate School of Systemic Neurosciences, Ludwig-Maximilians-University, Planegg 82152, Germany
| |
Collapse
|
21
|
Lemercier P, Vergallo A, Lista S, Zetterberg H, Blennow K, Potier MC, Habert MO, Lejeune FX, Dubois B, Teipel S, Hampel H. Association of plasma Aβ40/Aβ42 ratio and brain Aβ accumulation: testing a whole-brain PLS-VIP approach in individuals at risk of Alzheimer's disease. Neurobiol Aging 2021; 107:57-69. [PMID: 34388400 DOI: 10.1016/j.neurobiolaging.2021.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/25/2021] [Accepted: 07/07/2021] [Indexed: 11/29/2022]
Abstract
Molecular and brain regional/network-wise pathophysiological changes at preclinical stages of Alzheimer's disease (AD) have primarily been found through knowledge-based studies conducted in late-stage mild cognitive impairment/dementia populations. However, such an approach may compromise the objective of identifying the earliest spatial-temporal pathophysiological processes. We investigated 261 individuals with subjective memory complaints, a condition at increased risk of AD, to test a whole-brain, non-a-priori method based on partial least squares in unraveling the association between plasma Aβ42/Aβ40 ratio and an extensive set of brain regions characterized through molecular imaging of Aβ accumulation and cortical metabolism. Significant associations were mapped onto large-scale networks, identified through an atlas and by knowledge, to elaborate on the reliability of the results. Plasma Aβ42/40 ratio was associated with Aβ-PET uptake (but not FDG-PET) in regions generally investigated in preclinical AD such as those belonging to the default mode network, but also in regions/networks normally not accounted - including the central executive and salience networks - which likely have a selective vulnerability to incipient Aβ accumulation. The present whole-brain approach is promising to investigate early pathophysiological changes of AD to fully capture the complexity of the disease, which is essential to develop timely screening, detection, diagnostic, and therapeutic interventions.
Collapse
Affiliation(s)
- Pablo Lemercier
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France.
| | - Andrea Vergallo
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
| | - Simone Lista
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden; Department of Neurodegenerative Disease, UCL Institute of Neurology, Queen Square, London, UK; UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience & Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden; Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Marie-Claude Potier
- ICM Institut du Cerveau et de la Moelle épinière, CNRS UMR7225, INSERM U1127, UPMC, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Marie-Odile Habert
- Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, Paris, France; Centre pour l'Acquisition et le Traitement des Images (www.cati-neuroimaging.com), Paris, France; Department of Psychosomatic Medicine, University Medicine Rostock, Rostock, Germany; AP-HP, Hôpital Pitié-Salpêtrière, Département de Médecine Nucléaire, Paris, France
| | - François-Xavier Lejeune
- Bioinformatics and Biostatistics Core Facility iCONICS, Sorbonne Université UMR S 1127, Institut du Cerveau et de La Moelle Épinière, Paris, France
| | - Bruno Dubois
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France
| | - Stefan Teipel
- Clinical Dementia Research Section, German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany
| | - Harald Hampel
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Boulevard de l'hôpital, Paris, France.
| | | |
Collapse
|
22
|
Zorzi G, Cecchin D, Bussè C, Perini G, Corbetta M, Cagnin A. Changes of Metabolic Connectivity in Dementia with Lewy Bodies with Visual Hallucinations: A 18F-Fluorodeoxyglucose Positron Emission Tomography/Magnetic Resonance Study. Brain Connect 2021; 11:518-528. [PMID: 33757301 DOI: 10.1089/brain.2020.0988] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background: Recurrent complex visual hallucinations (VHs) are common in dementia with Lewy bodies (DLB). Previous investigations suggest that VHs are associated with connectivity changes within and between large scale networks involved in visual processing and attention. Aim: To examine more directly whether VH in DLB reflects direct changes in neuronal activity between cortical regions assessing metabolic connectivity with 18F-fluorodeoxyglucose (FDG)-positron emission tomography (PET)/magnetic resonance and graph theory. Methods: Twenty-six patients with probable DLB (13 VHs and 13 no-VHs; mean age: 72.9 ± 6.87 years vs. 70.2 ± 7.96 years) were enrolled. T1-weighted 3T-MR images and FDG-PET data were coacquired using an integrated PET/MR scanner. MR images defined cortical parcels of the Shaefer-Yeo atlas for multiple functional networks. We computed in each parcel the regional standardized-uptake-values (SUV) corrected for partial volume and normalized to the cerebellar cortex. Strength degree, clustering coefficient, characteristic path length, and hubs were analyzed with graph analysis. Results: The mean 18F-FDG-PET SUVr of parcels belonging to the visual and dorsal attention networks (DANs) were significantly lower in the VH group (p = 0.01). Metabolism in the right temporoparietal cortex correlated with VH severity (R = -0.58; p < 0.01). VH patients showed weaker metabolic connectivity in the parietal, temporal, and occipital cortex of the default mode network, DAN, and visual networks, but more robust connectivity in the right insula and orbitofrontal cortex. A lower global efficiency characterized the VH group, except for ventral attention network and limbic network. Conclusions: VHs in DLB correlate with lower glucose metabolism and weaker metabolic connectivity in the parietal-occipital cortex, but stronger connectivity in the limbic system. Impact statement This study shows that application of the graph theory to 18F-fluorodeoxyglucose-positron emission tomography data, commonly acquired during the diagnostic workflow in neurodegenerative diseases, could be used to obtain information of functional connectivity at a group level, with results that are consistent with other data commonly used in brain functional investigation (e.g., electroencephalography or functional magnetic resonance). New network-based methods of metabolic image analyses, such as graph analysis, are a recent area of research with a potential capacity to extract information on alterations of metabolic connectivity that may become pharmacological and neuromodulation targets of the physiopathology of recurrent complex visual hallucinations.
Collapse
Affiliation(s)
- Giovanni Zorzi
- Department of Neuroscience, University of Padova, Padova, Italy.,Padova Neuroscience Center, University of Padova, Padova, Italy
| | - Diego Cecchin
- Padova Neuroscience Center, University of Padova, Padova, Italy.,Nuclear Medicine Unit, Department of Medicine, University of Padova, Padova, Italy
| | - Cinzia Bussè
- Department of Neuroscience, University of Padova, Padova, Italy
| | | | - Maurizio Corbetta
- Department of Neuroscience, University of Padova, Padova, Italy.,Padova Neuroscience Center, University of Padova, Padova, Italy
| | - Annachiara Cagnin
- Department of Neuroscience, University of Padova, Padova, Italy.,Padova Neuroscience Center, University of Padova, Padova, Italy
| |
Collapse
|
23
|
A link between synaptic plasticity and reorganization of brain activity in Parkinson's disease. Proc Natl Acad Sci U S A 2021; 118:2013962118. [PMID: 33431672 PMCID: PMC7826364 DOI: 10.1073/pnas.2013962118] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The link between synaptic plasticity and reorganization of brain activity in health and disease remains a scientific challenge. We examined this question in Parkinson's disease (PD) where functional up-regulation of postsynaptic D2 receptors has been documented while its significance at the neural activity level has never been identified. We investigated cortico-subcortical plasticity in PD using the oculomotor system as a model to study reorganization of dopaminergic networks. This model is ideal because this system reorganizes due to frontal-to-parietal shifts in blood oxygen level-dependent (BOLD) activity. We tested the prediction that functional activation plasticity is associated with postsynaptic dopaminergic modifications by combining positron emission tomography/functional magnetic resonance imaging to investigate striatal postsynaptic reorganization of dopamine D2 receptors (using 11C-raclopride) and neural activation in PD. We used covariance (connectivity) statistics at molecular and functional levels to probe striato-cortical reorganization in PD in on/off medication states to show that functional and molecular forms of reorganization are related. D2 binding across regions defined by prosaccades showed increased molecular connectivity between both caudate/putamen and hyperactive parietal eye fields in PD in contrast with frontal eye fields in controls, in line with the shift model. Concerning antisaccades, parietal-striatal connectivity dominated in again in PD, unlike frontal regions. Concerning molecular-BOLD covariance, a striking sign reversal was observed: PD patients showed negative frontal-putamen functional-molecular associations, consistent with the reorganization shift, in contrast with the positive correlations observed in controls. Follow-up analysis in off-medication PD patients confirmed the negative BOLD-molecular correlation. These results provide a link among BOLD responses, striato-cortical synaptic reorganization, and neural plasticity in PD.
Collapse
|
24
|
Jamadar SD, Ward PGD, Liang EX, Orchard ER, Chen Z, Egan GF. Metabolic and Hemodynamic Resting-State Connectivity of the Human Brain: A High-Temporal Resolution Simultaneous BOLD-fMRI and FDG-fPET Multimodality Study. Cereb Cortex 2021; 31:2855-2867. [PMID: 33529320 DOI: 10.1093/cercor/bhaa393] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/26/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022] Open
Abstract
Simultaneous [18F]-fluorodeoxyglucose positron emission tomography functional magnetic resonance imaging (FDG-PET/fMRI) provides the capacity to image 2 sources of energetic dynamics in the brain-glucose metabolism and the hemodynamic response. fMRI connectivity has been enormously useful for characterizing interactions between distributed brain networks in humans. Metabolic connectivity based on static FDG-PET has been proposed as a biomarker for neurological disease, but FDG-sPET cannot be used to estimate subject-level measures of "connectivity," only across-subject "covariance." Here, we applied high-temporal resolution constant infusion functional positron emission tomography (fPET) to measure subject-level metabolic connectivity simultaneously with fMRI connectivity. fPET metabolic connectivity was characterized by frontoparietal connectivity within and between hemispheres. fPET metabolic connectivity showed moderate similarity with fMRI primarily in superior cortex and frontoparietal regions. Significantly, fPET metabolic connectivity showed little similarity with FDG-sPET metabolic covariance, indicating that metabolic brain connectivity is a nonergodic process whereby individual brain connectivity cannot be inferred from group-level metabolic covariance. Our results highlight the complementary strengths of fPET and fMRI in measuring the intrinsic connectivity of the brain and open up the opportunity for novel fundamental studies of human brain connectivity as well as multimodality biomarkers of neurological diseases.
Collapse
Affiliation(s)
- Sharna D Jamadar
- Monash Biomedical Imaging, Melbourne, Vic, 3800 Australia.,Turner Institute for Brain and Mental Health, Monash University, Melbourne, Vic, 3800 Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, Melbourne 3800, Australia
| | - Phillip G D Ward
- Monash Biomedical Imaging, Melbourne, Vic, 3800 Australia.,Turner Institute for Brain and Mental Health, Monash University, Melbourne, Vic, 3800 Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, Melbourne 3800, Australia
| | - Emma X Liang
- Monash Biomedical Imaging, Melbourne, Vic, 3800 Australia
| | - Edwina R Orchard
- Monash Biomedical Imaging, Melbourne, Vic, 3800 Australia.,Turner Institute for Brain and Mental Health, Monash University, Melbourne, Vic, 3800 Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, Melbourne 3800, Australia
| | - Zhaolin Chen
- Monash Biomedical Imaging, Melbourne, Vic, 3800 Australia.,Department of Electrical and Computer Systems Engineering, Monash University, Melbourne, Vic, 3800 Australia
| | - Gary F Egan
- Monash Biomedical Imaging, Melbourne, Vic, 3800 Australia.,Turner Institute for Brain and Mental Health, Monash University, Melbourne, Vic, 3800 Australia.,Australian Research Council Centre of Excellence for Integrative Brain Function, Melbourne 3800, Australia
| |
Collapse
|
25
|
Gonzalez-Escamilla G, Miederer I, Grothe MJ, Schreckenberger M, Muthuraman M, Groppa S. Metabolic and amyloid PET network reorganization in Alzheimer's disease: differential patterns and partial volume effects. Brain Imaging Behav 2021; 15:190-204. [PMID: 32125613 PMCID: PMC7835313 DOI: 10.1007/s11682-019-00247-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder, considered a disconnection syndrome with regional molecular pattern abnormalities quantifiable by the aid of PET imaging. Solutions for accurate quantification of network dysfunction are scarce. We evaluate the extent to which PET molecular markers reflect quantifiable network metrics derived through the graph theory framework and how partial volume effects (PVE)-correction (PVEc) affects these PET-derived metrics 75 AD patients and 126 cognitively normal older subjects (CN). Therefore our goal is twofold: 1) to evaluate the differential patterns of [18F]FDG- and [18F]AV45-PET data to depict AD pathology; and ii) to analyse the effects of PVEc on global uptake measures of [18F]FDG- and [18F]AV45-PET data and their derived covariance network reconstructions for differentiating between patients and normal older subjects. Network organization patterns were assessed using graph theory in terms of “degree”, “modularity”, and “efficiency”. PVEc evidenced effects on global uptake measures that are specific to either [18F]FDG- or [18F]AV45-PET, leading to increased statistical differences between the groups. PVEc was further shown to influence the topological characterization of PET-derived covariance brain networks, leading to an optimised characterization of network efficiency and modularisation. Partial-volume effects correction improves the interpretability of PET data in AD and leads to optimised characterization of network properties for organisation or disconnection.
Collapse
Affiliation(s)
- Gabriel Gonzalez-Escamilla
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Isabelle Miederer
- Department of Nuclear Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Michel J Grothe
- German Center for Neurodegenerative Diseases (DZNE) - Rostock/Greifswald, Rostock, Germany
| | - Mathias Schreckenberger
- Department of Nuclear Medicine, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Muthuraman Muthuraman
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Sergiu Groppa
- Department of Neurology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | | |
Collapse
|
26
|
Carli G, Tondo G, Boccalini C, Perani D. Brain Molecular Connectivity in Neurodegenerative Conditions. Brain Sci 2021; 11:brainsci11040433. [PMID: 33800680 PMCID: PMC8067093 DOI: 10.3390/brainsci11040433] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/15/2021] [Accepted: 03/23/2021] [Indexed: 12/28/2022] Open
Abstract
Positron emission tomography (PET) allows for the in vivo assessment of early brain functional and molecular changes in neurodegenerative conditions, representing a unique tool in the diagnostic workup. The increased use of multivariate PET imaging analysis approaches has provided the chance to investigate regional molecular processes and long-distance brain circuit functional interactions in the last decade. PET metabolic and neurotransmission connectome can reveal brain region interactions. This review is an overview of concepts and methods for PET molecular and metabolic covariance assessment with evidence in neurodegenerative conditions, including Alzheimer’s disease and Lewy bodies disease spectrum. We highlight the effects of environmental and biological factors on brain network organization. All of the above might contribute to innovative diagnostic tools and potential disease-modifying interventions.
Collapse
Affiliation(s)
- Giulia Carli
- School of Psychology, Vita-Salute San Raffaele University, 20121 Milan, Italy; (G.C.); (G.T.); (C.B.)
- In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20121 Milan, Italy
| | - Giacomo Tondo
- School of Psychology, Vita-Salute San Raffaele University, 20121 Milan, Italy; (G.C.); (G.T.); (C.B.)
- In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20121 Milan, Italy
| | - Cecilia Boccalini
- School of Psychology, Vita-Salute San Raffaele University, 20121 Milan, Italy; (G.C.); (G.T.); (C.B.)
- In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20121 Milan, Italy
| | - Daniela Perani
- School of Psychology, Vita-Salute San Raffaele University, 20121 Milan, Italy; (G.C.); (G.T.); (C.B.)
- In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20121 Milan, Italy
- Nuclear Medicine Unit, San Raffaele Hospital, 20121 Milan, Italy
- Correspondence: ; Tel.: +39-02-26432224
| |
Collapse
|
27
|
Boccalini C, Carli G, Pilotto A, Padovani A, Perani D. Gender-Related Vulnerability of Dopaminergic Neural Networks in Parkinson's Disease. Brain Connect 2020; 11:3-11. [PMID: 33198485 DOI: 10.1089/brain.2020.0781] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Background: In Parkinson's disease (PD), neurodegeneration of dopaminergic systems leads to motor and non-motor abnormalities. Sex might influence the clinical PD phenotypes and progression. Previous molecular imaging data focused only on the nigro-striato-cortical dopamine system that appeared more preserved in women. There is still a lack of evidence on gender/sex differences in the mesolimbic dopaminergic system. We aimed at assessing PD gender differences in both the dopaminergic pathways, by using a brain metabolic connectivity approach. This is based on the evidence of a significant coupling between the neurotransmission and metabolic impairments. Methods: We included 34 idiopathic PD patients (Female/Male: 16/18) and 34 healthy controls for comparison. The molecular architecture of both the dopaminergic networks was estimated throughout partial correlation analyses using brain metabolism data obtained by fluorine-18-fluorodeoxyglucose positron emission tomography (threshold set at p < 0.01, corrected for Bonferroni multiple comparisons). Results: Male patients were characterized by a widespread altered connectivity in the nigro-striato-cortical network and a sparing of the mesolimbic pathway. On the contrary, PD females showed a severe altered connectivity in the mesolimbic network and only a partial reconfiguration of the nigro-striato-cortical network. Discussion: Our findings add remarkable knowledge on the neurobiology of gender differences in PD, with the identification of specific neural vulnerabilities. The gender differences here revealed might be due to the combination of both biological and sociodemographic life factors. Gender differences in PD should be considered also for treatments and the targeting of modifiable risk factors.
Collapse
Affiliation(s)
- Cecilia Boccalini
- Vita-Salute San Raffaele University, Milan, Italy.,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulia Carli
- Vita-Salute San Raffaele University, Milan, Italy.,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Parkinson's Disease Rehabilitation Centre, FERB ONLUS S. Isidoro Hospital, Trescore, Italy
| | - Alessandro Padovani
- Parkinson's Disease Rehabilitation Centre, FERB ONLUS S. Isidoro Hospital, Trescore, Italy
| | - Daniela Perani
- Vita-Salute San Raffaele University, Milan, Italy.,In Vivo Human Molecular and Structural Neuroimaging Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Nuclear Medicine Unit, San Raffaele Hospital, Milan, Italy
| |
Collapse
|
28
|
Mihaescu AS, Kim J, Masellis M, Graff-Guerrero A, Cho SS, Christopher L, Valli M, Díez-Cirarda M, Koshimori Y, Strafella AP. Graph theory analysis of the dopamine D2 receptor network in Parkinson's disease patients with cognitive decline. J Neurosci Res 2020; 99:947-965. [PMID: 33271630 DOI: 10.1002/jnr.24760] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 11/14/2020] [Indexed: 12/30/2022]
Abstract
Cognitive decline in Parkinson's disease (PD) is a common sequela of the disorder that has a large impact on patient well-being. Its physiological etiology, however, remains elusive. Our study used graph theory analysis to investigate the large-scale topological patterns of the extrastriatal dopamine D2 receptor network. We used positron emission tomography with [11 C]FLB-457 to measure the binding potential of cortical dopamine D2 receptors in two networks: the meso-cortical dopamine network and the meso-limbic dopamine network. We also investigated the application of partial volume effect correction (PVEC) in conjunction with graph theory analysis. Three groups were investigated in this study divided according to their cognitive status as measured by the Montreal Cognitive Assessment score, with a score ≤25 considered cognitively impaired: (a) healthy controls (n = 13, 11 female), (b) cognitively unimpaired PD patients (PD-CU, n = 13, 5 female), and (c) PD patients with mild cognitive impairment (PD-MCI, n = 17, 4 female). In the meso-cortical network, we observed increased small-worldness, normalized clustering, and local efficiency in the PD-CU group compared to the PD-MCI group, as well as a hub shift in the PD-MCI group. Compensatory reorganization of the meso-cortical dopamine D2 receptor network may be responsible for some of the cognitive preservation observed in PD-CU. These results were found without PVEC applied and PVEC proved detrimental to the graph theory analysis. Overall, our findings demonstrate how graph theory analysis can be used to detect subtle changes in the brain that would otherwise be missed by regional comparisons of receptor density.
Collapse
Affiliation(s)
- Alexander S Mihaescu
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada.,Division of Brain, Imaging and Behaviour - Systems Neuroscience, Krembil Research Institute, University Health Network, University of Toronto, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, ON, Canada
| | - Jinhee Kim
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada
| | - Mario Masellis
- Institute of Medical Science, University of Toronto, ON, Canada.,LC Campbell Cognitive Neurology Research Unit, Sunnybrook Research Institute, University of Toronto, Toronto, ON, Canada.,Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Ariel Graff-Guerrero
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, ON, Canada
| | - Sang Soo Cho
- Division of Brain, Imaging and Behaviour - Systems Neuroscience, Krembil Research Institute, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Leigh Christopher
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada
| | - Mikaeel Valli
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada.,Division of Brain, Imaging and Behaviour - Systems Neuroscience, Krembil Research Institute, University Health Network, University of Toronto, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, ON, Canada
| | - María Díez-Cirarda
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada.,Division of Brain, Imaging and Behaviour - Systems Neuroscience, Krembil Research Institute, University Health Network, University of Toronto, Toronto, ON, Canada.,Neurodegenerative Diseases Group, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Yuko Koshimori
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada
| | - Antonio P Strafella
- Research Imaging Centre, Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, University of Toronto, Toronto, ON, Canada.,Division of Brain, Imaging and Behaviour - Systems Neuroscience, Krembil Research Institute, University Health Network, University of Toronto, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, ON, Canada.,Morton and Gloria Shulman Movement Disorder Unit & E.J. Safra Program in Parkinson Disease, Neurology Division, Department of Medicine, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
29
|
Carli G, Caminiti SP, Sala A, Galbiati A, Pilotto A, Ferini-Strambi L, Padovani A, Perani D. Impaired metabolic brain networks associated with neurotransmission systems in the α-synuclein spectrum. Parkinsonism Relat Disord 2020; 81:113-122. [DOI: 10.1016/j.parkreldis.2020.10.036] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/13/2020] [Accepted: 10/20/2020] [Indexed: 01/31/2023]
|
30
|
Shim HK, Lee HJ, Kim SE, Lee BI, Park S, Park KM. Alterations in the metabolic networks of temporal lobe epilepsy patients: A graph theoretical analysis using FDG-PET. Neuroimage Clin 2020; 27:102349. [PMID: 32702626 PMCID: PMC7374556 DOI: 10.1016/j.nicl.2020.102349] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 07/10/2020] [Accepted: 07/12/2020] [Indexed: 12/22/2022]
Abstract
OBJECTIVE The aim of this study is to investigate changes in metabolic networks based on fluorodeoxyglucose positron emission tomography (FDG-PET) in patients with drug-resistant temporal lobe epilepsy (TLE) (with and without hippocampal sclerosis [HS]) when compared with healthy controls. METHODS We retrospectively enrolled 30 patients with drug-resistant temporal lobe epilepsy (17 patients with HS and 13 patients without HS) and 39 healthy controls. All subjects underwent interictal FDG-PET scans, which were analyzed to obtain metabolic connectivity using graph theoretical analysis. We investigated the differences in metabolic connectivity between patients with drug-resistant TLE (with and without HS) and healthy controls. RESULTS When compared with healthy controls, TLE patients with HS showed alterations of global and local metabolic connectivity. When considering global connectivity, TLE patients with HS had a decreased average degree with increased modularity. When considering local connectivity, TLE patients with HS displayed alterations of betweeness centrality in widespread regions. However, there were no alterations of global metabolic connectivity in TLE patients without HS when compared with healthy controls. In addition, when compared to TLE patients without HS, TLE patients with HS had increased modularity. SIGNIFICANCE Our study demonstrates more severe alterations in metabolic networks based on FDG-PET in TLE patients with HS than in those without HS and healthy controls. This may represent distinct epileptic networks in TLE patients with HS versus those without HS, although both are drug-resistant focal epilepsy.
Collapse
Affiliation(s)
- Hye-Kyung Shim
- Department of Nuclear Medicine, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Ho-Joon Lee
- Department of Radiology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Sung Eun Kim
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Byung In Lee
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Seongho Park
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Kang Min Park
- Department of Neurology, Haeundae Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea.
| |
Collapse
|
31
|
Rahmani F, Sanjari Moghaddam H, Rahmani M, Aarabi MH. Metabolic connectivity in Alzheimer’s diseases. Clin Transl Imaging 2020. [DOI: 10.1007/s40336-020-00371-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
32
|
Hanekamp S, Simonyan K. The large-scale structural connectome of task-specific focal dystonia. Hum Brain Mapp 2020; 41:3253-3265. [PMID: 32311207 PMCID: PMC7375103 DOI: 10.1002/hbm.25012] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/27/2020] [Accepted: 04/06/2020] [Indexed: 12/19/2022] Open
Abstract
The emerging view of dystonia is that of a large‐scale functional network disorder, in which the communication is disrupted between sensorimotor cortical areas, basal ganglia, thalamus, and cerebellum. The structural underpinnings of functional alterations in dystonia are, however, poorly understood. Notably, it is unclear whether structural changes form a larger‐scale dystonic network or rather remain focal to isolated brain regions, merely underlying their functional abnormalities. Using diffusion‐weighted imaging and graph theoretical analysis, we examined inter‐regional white matter connectivity of the whole‐brain structural network in two different forms of task‐specific focal dystonia, writer's cramp and laryngeal dystonia, compared to healthy individuals. We show that, in addition to profoundly altered functional network in focal dystonia, its structural connectome is characterized by large‐scale aberrations due to abnormal transfer of prefrontal and parietal nodes between neural communities and the reorganization of normal hub architecture, commonly involving the insula and superior frontal gyrus in patients compared to controls. Other prominent common changes involved the basal ganglia, parietal and cingulate cortical regions, whereas premotor and occipital abnormalities distinctly characterized the two forms of dystonia. We propose a revised pathophysiological model of focal dystonia as a disorder of both functional and structural connectomes, where dystonia form‐specific abnormalities underlie the divergent mechanisms in the development of distinct clinical symptomatology. These findings may guide the development of novel therapeutic strategies directed at targeted neuromodulation of pathophysiological brain regions for the restoration of their structural and functional connectivity.
Collapse
Affiliation(s)
- Sandra Hanekamp
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA.,Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, USA
| | - Kristina Simonyan
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA.,Department of Otolaryngology-Head and Neck Surgery, Harvard Medical School, Boston, Massachusetts, USA.,Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
33
|
Verger A, Horowitz T, Chawki MB, Eusebio A, Bordonne M, Azulay JP, Girard N, Guedj E. From metabolic connectivity to molecular connectivity: application to dopaminergic pathways. Eur J Nucl Med Mol Imaging 2019; 47:413-424. [DOI: 10.1007/s00259-019-04574-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 10/10/2019] [Indexed: 12/11/2022]
|
34
|
Ashok AH, Myers J, Reis Marques T, Rabiner EA, Howes OD. Reduced mu opioid receptor availability in schizophrenia revealed with [ 11C]-carfentanil positron emission tomographic Imaging. Nat Commun 2019; 10:4493. [PMID: 31582737 PMCID: PMC6776653 DOI: 10.1038/s41467-019-12366-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 09/04/2019] [Indexed: 12/16/2022] Open
Abstract
Negative symptoms, such as amotivation and anhedonia, are a major cause of functional impairment in schizophrenia. There are currently no licensed treatments for negative symptoms, highlighting the need to understand the molecular mechanisms underlying them. Mu-opioid receptors (MOR) in the striatum play a key role in hedonic processing and reward function and are reduced post-mortem in schizophrenia. However, it is unknown if mu-opioid receptor availability is altered in-vivo or related to negative symptoms in schizophrenia. Using [11 C]-carfentanil positron emission tomography (PET) scans in 19 schizophrenia patients and 20 age-matched healthy controls, here we show a significantly lower MOR availability in patients with schizophrenia in the striatum (Cohen's d = 0.7), and the hedonic network. In addition, we report a marked global increase in inter-regional covariance of MOR availability in schizophrenia, largely due to increased cortical-subcortical covariance.
Collapse
Affiliation(s)
- Abhishekh H Ashok
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Hammersmith Hospital, Imperial College London, London, UK
- Faculty of Medicine, Psychiatric Imaging Group, Institute of Clinical Sciences (ICS), Imperial College London, London, UK
- Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Jim Myers
- Faculty of Medicine, Imperial College London, London, UK
| | - Tiago Reis Marques
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Hammersmith Hospital, Imperial College London, London, UK
- Faculty of Medicine, Psychiatric Imaging Group, Institute of Clinical Sciences (ICS), Imperial College London, London, UK
- Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Eugenii A Rabiner
- Invicro, London, UK
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK
| | - Oliver D Howes
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Hammersmith Hospital, Imperial College London, London, UK.
- Faculty of Medicine, Psychiatric Imaging Group, Institute of Clinical Sciences (ICS), Imperial College London, London, UK.
- Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, UK.
| |
Collapse
|
35
|
Sala A, Perani D. Brain Molecular Connectivity in Neurodegenerative Diseases: Recent Advances and New Perspectives Using Positron Emission Tomography. Front Neurosci 2019; 13:617. [PMID: 31258466 PMCID: PMC6587303 DOI: 10.3389/fnins.2019.00617] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/29/2019] [Indexed: 12/12/2022] Open
Abstract
Positron emission tomography (PET) represents a unique molecular tool to get in vivo access to a wide spectrum of biological and neuropathological processes, of crucial relevance for neurodegenerative conditions. Although most PET findings are based on massive univariate approaches, in the last decade the increasing interest in multivariate methods has paved the way to the assessment of unexplored cerebral features, spanning from resting state brain networks to whole-brain connectome properties. Currently, the combination of molecular neuroimaging techniques with multivariate connectivity methods represents one of the most powerful, yet still emerging, approach to achieve novel insights into the pathophysiology of neurodegenerative diseases. In this review, we will summarize the available evidence in the field of PET molecular connectivity, with the aim to provide an overview of how these studies may increase the understanding of the pathogenesis of neurodegenerative diseases, over and above "traditional" structural/functional connectivity studies. Considering the available evidence, a major focus will be represented by molecular connectivity studies using [18F]FDG-PET, today applied in the major neuropathological spectra, from amyloidopathies and tauopathies to synucleinopathies and beyond. Pioneering studies using PET tracers targeting brain neuropathology and neurotransmission systems for connectivity studies will be discussed, their strengths and limitations highlighted with reference to both applied methodology and results interpretation. The most common methods for molecular connectivity assessment will be reviewed, with particular emphasis on the available strategies to investigate molecular connectivity at the single-subject level, of potential relevance for not only research but also diagnostic purposes. Finally, we will highlight possible future perspectives in the field, with reference in particular to newly available PET tracers, which will expand the application of molecular connectivity to new, exciting, unforeseen possibilities.
Collapse
Affiliation(s)
- Arianna Sala
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, Milan, Italy.,Division of Neuroscience, Faculty of Psychology, San Raffaele Scientific Institute (IRCCS), Milan, Italy
| | - Daniela Perani
- Division of Neuroscience, Faculty of Psychology, San Raffaele Scientific Institute (IRCCS), Milan, Italy.,Faculty of Psychology, Vita-Salute San Raffaele University, Milan, Italy.,Nuclear Medicine Unit, Faculty of Psychology, San Raffaele Hospital (IRCCS), Milan, Italy
| |
Collapse
|
36
|
Head-to-head comparison of 11C-PBR28 and 11C-ER176 for quantification of the translocator protein in the human brain. Eur J Nucl Med Mol Imaging 2019; 46:1822-1829. [DOI: 10.1007/s00259-019-04349-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/29/2019] [Indexed: 10/26/2022]
|