1
|
Lagache L, Zirem Y, Le Rhun É, Fournier I, Salzet M. Heterogeneity Assessment and Protein Pathway Prediction via Spatial Lipidomic and Proteomic Correlation: Advancing Dry Proteomics concept for Human Glioblastoma Prognosis. Mol Cell Proteomics 2024:100891. [PMID: 39644924 DOI: 10.1016/j.mcpro.2024.100891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 11/20/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024] Open
Abstract
Prediction of proteins and associated biological pathways from lipid analyses via MALDI MSI is a pressing challenge. We introduced "dry proteomics," using MALDI MSI to validate spatial localization of identified optimal clusters in lipid imaging. Consistent cluster appearance across omics images suggests association with specific lipid and protein in distinct biological pathways, forming the basis of dry proteomics. The methodology was refined using rat brain tissue as a model, then applied to human glioblastoma, a highly heterogeneous cancer. Sequential tissue sections underwent omics MALDI MSI and unsupervised clustering. Spatial omics analysis facilitated lipid and protein characterization, leading to a predictive model identifying clusters in any tissue based on unique lipid signatures and predicting associated protein pathways. Application to rat brain slices revealed diverse tissue subpopulations, including successfully predicted cerebellum areas. Similarly, the methodology was applied to a dataset from a cohort of 50 glioblastoma patients, reused from a previous study. However, among the 50 patients, only 13 lipid signatures from MALDI MSI data were available, allowing for the identification of lipid-protein associations that correlated with patient prognosis. For cases lacking lipid imaging data, a classification model based on protein data was developed from dry proteomic results to effectively categorize the remaining cohort.
Collapse
Affiliation(s)
- Laurine Lagache
- Univ.Lille, Inserm, CHU Lille, U1192 - Proteomics Inflammatory Response Mass Spectrometry- PRISM, F-59000 Lille, France
| | - Yanis Zirem
- Univ.Lille, Inserm, CHU Lille, U1192 - Proteomics Inflammatory Response Mass Spectrometry- PRISM, F-59000 Lille, France
| | - Émilie Le Rhun
- Univ.Lille, Inserm, CHU Lille, U1192 - Proteomics Inflammatory Response Mass Spectrometry- PRISM, F-59000 Lille, France; Department of Neurosurgery and Neurology, Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Isabelle Fournier
- Univ.Lille, Inserm, CHU Lille, U1192 - Proteomics Inflammatory Response Mass Spectrometry- PRISM, F-59000 Lille, France; Institut Universitaire de France, 75000 Paris.
| | - Michel Salzet
- Univ.Lille, Inserm, CHU Lille, U1192 - Proteomics Inflammatory Response Mass Spectrometry- PRISM, F-59000 Lille, France; Institut Universitaire de France, 75000 Paris.
| |
Collapse
|
2
|
Shao Q, Li Y, Jin L, Zhou S, Fu X, Liu T, Luo G, Du S, Chen C. Semen Cuscutae flavonoids activated the cAMP-PKA-CREB-BDNF pathway and exerted an antidepressant effect in mice. Front Pharmacol 2024; 15:1491900. [PMID: 39654620 PMCID: PMC11625582 DOI: 10.3389/fphar.2024.1491900] [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: 09/06/2024] [Accepted: 11/06/2024] [Indexed: 12/12/2024] Open
Abstract
Background Semen Cuscutae flavonoids (SCFs) constitute a class of metabolites of Semen Cuscutae, a botanical drug that was recently found to have an anti-depression effect. This study aimed to evaluate the anti-depression effects of SCFs in chronic unpredictable mild stress (CUMS)-induced mice and to interrogate the underlying mechanisms. Materials and methods The CUMS mice were used for assessing the effects of SCFs treatments on depression. Mice were randomly divided into five groups. Four groups were subjected to the CUMS induction and concomitantly administered orally with either the vehicle or with a high-, medium-, and low-dose of SCFs, once per day for 4 weeks. One group was kept untreated as a control. The mice were then assessed for their statuses of a number of depression-related parameters, including body weight, food intake, sucrose preference test (SPT), open field test (OFT), tail suspension test (TST), and forced swim test (FST). In addition, a day after the completion of these tests, biopsies from the hippocampus were harvested and used to perform metabolomics by HPLC-MS/MS and to assess the levels of cAMP by ELISA and the levels of PKA, CREB, p-CREB, and BDNF by Western blot analyses. Results SCFs resulted in significant increases in both body weight and food intake and in the amelioration of the depressive-like behaviors in CUMS mice. A high-dose SCFs treatment led to significant alterations in 72 metabolites, of which 26 were identified as potential biomarkers for the SCFs treatment. These metabolites are associated with lipid, amino acid, and nucleotide metabolism. Among 26 metabolites, cAMP was positively correlated with body weight, SPT, OFT-total distance, and OFT-central residence time, while negatively correlated with immobility time in TST and FST, linking a change in cAMP with the SCFs treatment and the significant improvement in depressive symptoms in CUMS mice. Further analyses revealed that the levels of cAMP, PKA, CREB, p-CREB, and BDNF were reduced in the hippocampus of CUMS mice but were all increased following the SCFs treatments. Conclusion SCFs could ameliorate hippocampal metabolic disturbances and depressive behaviors and cause the activation of the cAMP-PKA-CREB-BDNF signaling pathway in the hippocampus of CUMS mice.
Collapse
Affiliation(s)
- Qianfeng Shao
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yue Li
- Centre for Translational Medicine, Shenzhen Bao’an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Lin Jin
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Sheng Zhou
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaowei Fu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tong Liu
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Guangbin Luo
- Centre for Translational Medicine, Shenzhen Bao’an Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, China
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Shaohui Du
- Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Che Chen
- School of Traditional Chinese Medicine, Ningxia Medical University, Yinchuan, China
| |
Collapse
|
3
|
Shi X, Zhang X, Ao JF, Yang M. Correlation between Non-HDL-C/HDL-C and Aβ1-42 levels in cerebral infarction-related cognitive dysfunction. Clin Neurol Neurosurg 2024; 245:108503. [PMID: 39178633 DOI: 10.1016/j.clineuro.2024.108503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 08/26/2024]
Abstract
OBJECTIVE Cerebral infarction treatments are most effective if used early after stroke symptoms occur. Also, early detection is crucial for delaying and improving cognitive impairment. This study investigated the relationship between the ratio of non-high-density lipoprotein cholesterol to high-density lipoprotein cholesterol (Non-HDL-C/HDL-C), which reflects the entire burden of the cholesterol transported in atherogenic lipoproteins, and the level of β-amyloid 1-42 (Aβ-1-42), a major component of cerebrovascular amyloid deposits, in peripheral blood and cognitive dysfunction secondary to cerebral infarction. METHODS A total of 83 patients with cerebral infarction admitted to Bozhou People's Hospital between June 2019 and June 2022 were assessed. The patients were divided into two groups based on their Mini-Mental State Scale (MMSE) scores: cognitive dysfunction group (n = 30) and non-cognitive dysfunction group (n = 53). In addition, a control group comprising 34 patients with transient cerebral insufficiency or cerebrovascular stenosis was selected. The groups were compared in terms of various clinical factors, including gender, age, hypertension, hyperlipidemia, lipid indexes, Non-HDL-C/HDL-C, and Aβ1-42 levels. Logistic regression analysis was used to identify the risk factors associated with cognitive dysfunction. RESULTS The results showed that hypertensive patients with cognitive dysfunction secondary to cerebral infarction had a higher proportion of frontal lobe, temporal lobe, and thalamus involvement and lower scores on the MMSE compared to the non-cognitive impairment group and control group (p < 0.05). Additionally, the levels of homocysteine (HCY), Non-HDL-C/HDL-C, and Aβ1-42 in peripheral blood were significantly higher in hypertensive patients with cognitive dysfunction compared to the other two groups (all p < 0.05) and were identified as risk factors for cognitive dysfunction secondary to cerebral infarction. Peripheral blood levels of Non-HDL-C/HDL-C and Aβ1-42 are risk factors for secondary cognitive dysfunction following a cerebral infarction. CONCLUSION These data have important clinical implications for understanding the mechanisms underlying cognitive dysfunction in individuals with cerebrovascular disorders, potentially leading to new early interventions for preventing or treating such diseases.
Collapse
Affiliation(s)
- Xiufang Shi
- The Clinical Laboratory of Bozhou People's Hospital, BoZhou 236800, China.
| | - Xiaoqing Zhang
- The Clinical Laboratory of Bozhou People's Hospital, BoZhou 236800, China.
| | - Jia-Fu Ao
- The Clinical Laboratory of Bozhou People's Hospital, BoZhou 236800, China.
| | - Miao Yang
- The Clinical Laboratory of Bozhou People's Hospital, BoZhou 236800, China.
| |
Collapse
|
4
|
Mohamed Yusoff AA, Mohd Khair SZN. Unraveling mitochondrial dysfunction: comprehensive perspectives on its impact on neurodegenerative diseases. Rev Neurosci 2024:revneuro-2024-0080. [PMID: 39174305 DOI: 10.1515/revneuro-2024-0080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 07/30/2024] [Indexed: 08/24/2024]
Abstract
Neurodegenerative diseases represent a significant challenge to modern medicine, with their complex etiology and progressive nature posing hurdles to effective treatment strategies. Among the various contributing factors, mitochondrial dysfunction has emerged as a pivotal player in the pathogenesis of several neurodegenerative disorders. This review paper provides a comprehensive overview of how mitochondrial impairment contributes to the development of neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis, driven by bioenergetic defects, biogenesis impairment, alterations in mitochondrial dynamics (such as fusion or fission), disruptions in calcium buffering, lipid metabolism dysregulation and mitophagy dysfunction. It also covers current therapeutic interventions targeting mitochondrial dysfunction in these diseases.
Collapse
Affiliation(s)
- Abdul Aziz Mohamed Yusoff
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Siti Zulaikha Nashwa Mohd Khair
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
5
|
Tripp BA, Dillon ST, Yuan M, Asara JM, Vasunilashorn SM, Fong TG, Inouye SK, Ngo LH, Marcantonio ER, Xie Z, Libermann TA, Otu HH. Integrated Multi-Omics Analysis of Cerebrospinal Fluid in Postoperative Delirium. Biomolecules 2024; 14:924. [PMID: 39199312 PMCID: PMC11352186 DOI: 10.3390/biom14080924] [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: 06/29/2024] [Revised: 07/26/2024] [Accepted: 07/27/2024] [Indexed: 09/01/2024] Open
Abstract
Preoperative risk biomarkers for delirium may aid in identifying high-risk patients and developing intervention therapies, which would minimize the health and economic burden of postoperative delirium. Previous studies have typically used single omics approaches to identify such biomarkers. Preoperative cerebrospinal fluid (CSF) from the Healthier Postoperative Recovery study of adults ≥ 63 years old undergoing elective major orthopedic surgery was used in a matched pair delirium case-no delirium control design. We performed metabolomics and lipidomics, which were combined with our previously reported proteomics results on the same samples. Differential expression, clustering, classification, and systems biology analyses were applied to individual and combined omics datasets. Probabilistic graph models were used to identify an integrated multi-omics interaction network, which included clusters of heterogeneous omics interactions among lipids, metabolites, and proteins. The combined multi-omics signature of 25 molecules attained an AUC of 0.96 [95% CI: 0.85-1.00], showing improvement over individual omics-based classification. We conclude that multi-omics integration of preoperative CSF identifies potential risk markers for delirium and generates new insights into the complex pathways associated with delirium. With future validation, this hypotheses-generating study may serve to build robust biomarkers for delirium and improve our understanding of its pathophysiology.
Collapse
Affiliation(s)
- Bridget A. Tripp
- Department of Electrical and Computer Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Simon T. Dillon
- Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (S.T.D.)
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
| | - Min Yuan
- Division of Signal Transduction and Mass Spectrometry Core, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - John M. Asara
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Division of Signal Transduction and Mass Spectrometry Core, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Sarinnapha M. Vasunilashorn
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
- Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Tamara G. Fong
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
- Aging Brain Center, Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA 02131, USA
| | - Sharon K. Inouye
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
- Aging Brain Center, Marcus Institute for Aging Research, Hebrew SeniorLife, Boston, MA 02131, USA
| | - Long H. Ngo
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
- Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Edward R. Marcantonio
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Zhongcong Xie
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Towia A. Libermann
- Genomics, Proteomics, Bioinformatics and Systems Biology Center, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; (S.T.D.)
- Harvard Medical School, Boston, MA 02215, USA; (J.M.A.); (L.H.N.); (Z.X.)
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Hasan H. Otu
- Department of Electrical and Computer Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| |
Collapse
|
6
|
Bhargava A, Knapp JD, Fiehn O, Neylan TC, Inslicht SS. An exploratory study on lipidomic profiles in a cohort of individuals with posttraumatic stress disorder. Sci Rep 2024; 14:15256. [PMID: 38956202 PMCID: PMC11219863 DOI: 10.1038/s41598-024-62971-7] [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: 02/21/2024] [Accepted: 05/23/2024] [Indexed: 07/04/2024] Open
Abstract
Posttraumatic stress disorder (PTSD) can develop after trauma exposure. Some studies report that women develop PTSD at twice the rate of men, despite greater trauma exposure in men. Lipids and their metabolites (lipidome) regulate a myriad of key biological processes and pathways such as membrane integrity, oxidative stress, and neuroinflammation in the brain by maintaining neuronal connectivity and homeostasis. In this study, we analyzed the lipidome of 40 adults with PTSD and 40 trauma-exposed non-PTSD individuals (n = 20/sex/condition; 19-39 years old). Plasma samples were analyzed for lipidomics using Quadrupole Time-of-Flight (QToF) mass spectrometry. Additionally, ~ 90 measures were collected, on sleep, and mental and physical health indices. Poorer sleep quality was associated with greater PTSD severity in both sexes. The lipidomics analysis identified a total of 348 quantifiable known lipid metabolites and 1951 lipid metabolites that are yet unknown; known metabolites were part of 13 lipid subclasses. After adjusting for BMI and sleep quality, in women with PTSD, only one lipid subclass, phosphatidylethanolamine (PE) was altered, whereas, in men with PTSD, 9 out of 13 subclasses were altered compared to non-PTSD women and men, respectively. Severe PTSD was associated with 22% and 5% of altered lipid metabolites in men and women, respectively. Of the changed metabolites, only 0.5% measures (2 PEs and cholesterol) were common between women and men with PTSD. Several sphingomyelins, PEs, ceramides, and triglycerides were increased in men with severe PTSD. The correlations between triglycerides and ceramide metabolites with cholesterol metabolites and systolic blood pressure were dependent upon sex and PTSD status. Alterations in triglycerides and ceramides are linked with cardiac health and metabolic function in humans. Thus, disturbed sleep and higher body mass may have contributed to changes in the lipidome found in PTSD.
Collapse
Affiliation(s)
- Aditi Bhargava
- Department of Obstetrics and Gynecology, Center for Reproductive Sciences, University of California San Francisco, San Francisco, CA, 94143, USA.
- Aseesa Inc., Hillsborough, CA, 94010, USA.
| | | | - Oliver Fiehn
- NIH West Coast Metabolomics Center, University of California Davis Genome Center, Davis, CA, 95616, USA
| | - Thomas C Neylan
- San Francisco VA Health Care System, 4150 Clement St. (116P), San Francisco, CA, 94121, USA
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Sabra S Inslicht
- San Francisco VA Health Care System, 4150 Clement St. (116P), San Francisco, CA, 94121, USA.
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
7
|
Niccolai E, Pedone M, Martinelli I, Nannini G, Baldi S, Simonini C, Di Gloria L, Zucchi E, Ramazzotti M, Spezia PG, Maggi F, Quaranta G, Masucci L, Bartolucci G, Stingo FC, Mandrioli J, Amedei A. Amyotrophic lateral sclerosis stratification: unveiling patterns with virome, inflammation, and metabolism molecules. J Neurol 2024; 271:4310-4325. [PMID: 38644373 PMCID: PMC11233352 DOI: 10.1007/s00415-024-12348-7] [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: 01/30/2024] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/23/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is an untreatable and clinically heterogeneous condition primarily affecting motor neurons. The ongoing quest for reliable biomarkers that mirror the disease status and progression has led to investigations that extend beyond motor neurons' pathology, encompassing broader systemic factors such as metabolism, immunity, and the microbiome. Our study contributes to this effort by examining the potential role of microbiome-related components, including viral elements, such as torque tenovirus (TTV), and various inflammatory factors, in ALS. In our analysis of serum samples from 100 ALS patients and 34 healthy controls (HC), we evaluated 14 cytokines, TTV DNA load, and 18 free fatty acids (FFA). We found that the evaluated variables are effective in differentiating ALS patients from healthy controls. In addition, our research identifies four unique patient clusters, each characterized by distinct biological profiles. Intriguingly, no correlations were found with site of onset, sex, progression rate, phenotype, or C9ORF72 expansion. A remarkable aspect of our findings is the discovery of a gender-specific relationship between levels of 2-ethylhexanoic acid and patient survival. In addition to contributing to the growing body of evidence suggesting altered peripheral immune responses in ALS, our exploratory research underscores metabolic diversity challenging conventional clinical classifications. If our exploratory findings are validated by further research, they could significantly impact disease understanding and patient care customization. Identifying groups based on biological profiles might aid in clustering patients with varying responses to treatments.
Collapse
Affiliation(s)
- Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Matteo Pedone
- Department of Statistics, Computer Science, Applications "G. Parenti", University of Florence, Florence, Italy
| | - Ilaria Martinelli
- Neurology Unit, Department of Neuroscience, Azienda Ospedaliero Universitaria Di Modena, Modena, Italy
| | - Giulia Nannini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Simone Baldi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Cecilia Simonini
- Neurology Unit, Department of Neuroscience, Azienda Ospedaliero Universitaria Di Modena, Modena, Italy
| | - Leandro Di Gloria
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Elisabetta Zucchi
- Neurology Unit, Department of Neuroscience, Azienda Ospedaliero Universitaria Di Modena, Modena, Italy
| | - Matteo Ramazzotti
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Pietro Giorgio Spezia
- Department of Translational Research, Retrovirus Center - University of Pisa, Pisa, Italy
| | - Fabrizio Maggi
- Laboratory of Virology, National Institute for Infectious Diseases Lazzaro Spallanzani - IRCCS, Rome, Italy
| | - Gianluca Quaranta
- Department of Laboratory and Infectious Sciences, A. Gemelli University Hospital IRCCS, Rome, Italy
| | - Luca Masucci
- Department of Laboratory and Infectious Sciences, A. Gemelli University Hospital IRCCS, Rome, Italy
| | - Gianluca Bartolucci
- Department of Neurosciences, Psychology, Drug Research and Child Health Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Florence, Italy
| | - Francesco Claudio Stingo
- Department of Statistics, Computer Science, Applications "G. Parenti", University of Florence, Florence, Italy
| | - Jessica Mandrioli
- Neurology Unit, Department of Neuroscience, Azienda Ospedaliero Universitaria Di Modena, Modena, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| |
Collapse
|
8
|
Mondal K, Del Mar NA, Gary AA, Grambergs RC, Yousuf M, Tahia F, Stephenson B, Stephenson DJ, Chalfant CE, Reiner A, Mandal N. Sphingolipid changes in mouse brain and plasma after mild traumatic brain injury at the acute phases. Lipids Health Dis 2024; 23:200. [PMID: 38937745 PMCID: PMC11209960 DOI: 10.1186/s12944-024-02186-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 06/14/2024] [Indexed: 06/29/2024] Open
Abstract
BACKGROUND Traumatic brain injury (TBI) causes neuroinflammation and can lead to long-term neurological dysfunction, even in cases of mild TBI (mTBI). Despite the substantial burden of this disease, the management of TBI is precluded by an incomplete understanding of its cellular mechanisms. Sphingolipids (SPL) and their metabolites have emerged as key orchestrators of biological processes related to tissue injury, neuroinflammation, and inflammation resolution. No study so far has investigated comprehensive sphingolipid profile changes immediately following TBI in animal models or human cases. In this study, sphingolipid metabolite composition was examined during the acute phases in brain tissue and plasma of mice following mTBI. METHODS Wildtype mice were exposed to air-blast-mediated mTBI, with blast exposure set at 50-psi on the left cranium and 0-psi designated as Sham. Sphingolipid profile was analyzed in brain tissue and plasma during the acute phases of 1, 3, and 7 days post-TBI via liquid-chromatography-mass spectrometry. Simultaneously, gene expression of sphingolipid metabolic markers within brain tissue was analyzed using quantitative reverse transcription-polymerase chain reaction. Significance (P-values) was determined by non-parametric t-test (Mann-Whitney test) and by Tukey's correction for multiple comparisons. RESULTS In post-TBI brain tissue, there was a significant elevation of 1) acid sphingomyelinase (aSMase) at 1- and 3-days, 2) neutral sphingomyelinase (nSMase) at 7-days, 3) ceramide-1-phosphate levels at 1 day, and 4) monohexosylceramide (MHC) and sphingosine at 7-days. Among individual species, the study found an increase in C18:0 and a decrease in C24:1 ceramides (Cer) at 1 day; an increase in C20:0 MHC at 3 days; decrease in MHC C18:0 and increase in MHC C24:1, sphingomyelins (SM) C18:0, and C24:0 at 7 days. Moreover, many sphingolipid metabolic genes were elevated at 1 day, followed by a reduction at 3 days and an absence at 7-days post-TBI. In post-TBI plasma, there was 1) a significant reduction in Cer and MHC C22:0, and an increase in MHC C16:0 at 1 day; 2) a very significant increase in long-chain Cer C24:1 accompanied by significant decreases in Cer C24:0 and C22:0 in MHC and SM at 3 days; and 3) a significant increase of C22:0 in all classes of SPL (Cer, MHC and SM) as well as a decrease in Cer C24:1, MHC C24:1 and MHC C24:0 at 7 days. CONCLUSIONS Alterations in sphingolipid metabolite composition, particularly sphingomyelinases and short-chain ceramides, may contribute to the induction and regulation of neuroinflammatory events in the early stages of TBI, suggesting potential targets for novel diagnostic, prognostic, and therapeutic strategies in the future.
Collapse
Affiliation(s)
- Koushik Mondal
- Department of Ophthalmology, The University of Tennessee Health Science Centre, Memphis, TN, 38163, USA
- Molecular Diagnostics Laboratory, Department of Basic & Translational Research, Saroj Gupta Cancer Centre & Research Institute, Kolkata, WB, 700 063, India
| | - Nobel A Del Mar
- Department of Ophthalmology, The University of Tennessee Health Science Centre, Memphis, TN, 38163, USA
| | - Ashlyn A Gary
- Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Richard C Grambergs
- Department of Ophthalmology, The University of Tennessee Health Science Centre, Memphis, TN, 38163, USA
| | - Mohd Yousuf
- Department of Ophthalmology, The University of Tennessee Health Science Centre, Memphis, TN, 38163, USA
| | - Faiza Tahia
- Department of Ophthalmology, The University of Tennessee Health Science Centre, Memphis, TN, 38163, USA
| | - Benjamin Stephenson
- Departments of Medicine and Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
| | - Daniel J Stephenson
- Departments of Medicine and Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
| | - Charles E Chalfant
- Departments of Medicine and Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, 22903, USA
- Research Service, Richmond VA Medical Center, Richmond, VA, 23298, USA
| | - Anton Reiner
- Department of Ophthalmology, The University of Tennessee Health Science Centre, Memphis, TN, 38163, USA
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Centre, Memphis, TN, 38163, USA
| | - Nawajes Mandal
- Department of Ophthalmology, The University of Tennessee Health Science Centre, Memphis, TN, 38163, USA.
- Department of Anatomy and Neurobiology, The University of Tennessee Health Science Centre, Memphis, TN, 38163, USA.
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Centre, Memphis, TN, 38163, USA.
- Memphis VA Medical Center, Memphis, TN, 38104, USA.
| |
Collapse
|
9
|
Kwon S, Park KS, Yoon KH. Regulator of Lipid Metabolism NHR-49 Mediates Pathogen Avoidance through Precise Control of Neuronal Activity. Cells 2024; 13:978. [PMID: 38891110 PMCID: PMC11172349 DOI: 10.3390/cells13110978] [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: 04/05/2024] [Revised: 05/24/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Precise control of neuronal activity is crucial for the proper functioning of neurons. How lipid homeostasis contributes to neuronal activity and how much of it is regulated by cells autonomously is unclear. In this study, we discovered that absence of the lipid regulator nhr-49, a functional ortholog of the peroxisome proliferator-activated receptor (PPAR) in Caenorhabditis elegans, resulted in defective pathogen avoidance behavior against Pseudomonas aeruginosa (PA14). Functional NHR-49 was required in the neurons, and more specifically, in a set of oxygen-sensing body cavity neurons, URX, AQR, and PQR. We found that lowering the neuronal activity of the body cavity neurons improved avoidance in nhr-49 mutants. Calcium imaging in URX neurons showed that nhr-49 mutants displayed longer-lasting calcium transients in response to an O2 upshift, suggesting that excess neuronal activity leads to avoidance defects. Cell-specific rescue of NHR-49 in the body cavity neurons was sufficient to improve pathogen avoidance, as well as URX neuron calcium kinetics. Supplementation with oleic acid also improved avoidance behavior and URX calcium kinetics, suggesting that the defective calcium response in the neuron is due to lipid dysfunction. These findings highlight the role of cell-autonomous lipid regulation in neuronal physiology and immune behavior.
Collapse
Affiliation(s)
- Saebom Kwon
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea;
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
| | - Kyu-Sang Park
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea;
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
- Department of Global Medical Science, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
| | - Kyoung-hye Yoon
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju 26426, Republic of Korea
| |
Collapse
|
10
|
Ali MY, Bar-Peled L. Chemical proteomics to study metabolism, a reductionist approach applied at the systems level. Cell Chem Biol 2024; 31:446-451. [PMID: 38518745 DOI: 10.1016/j.chembiol.2024.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 11/02/2023] [Accepted: 02/28/2024] [Indexed: 03/24/2024]
Abstract
Cellular metabolism encompasses a complex array of interconnected biochemical pathways that are required for cellular homeostasis. When dysregulated, metabolism underlies multiple human pathologies. At the heart of metabolic networks are enzymes that have been historically studied through a reductionist lens, and more recently, using high throughput approaches including genomics and proteomics. Merging these two divergent viewpoints are chemical proteomic technologies, including activity-based protein profiling, which combines chemical probes specific to distinct enzyme families or amino acid residues with proteomic analysis. This enables the study of metabolism at the network level with the precision of powerful biochemical approaches. Herein, we provide a primer on how chemical proteomic technologies custom-built for studying metabolism have unearthed fundamental principles in metabolic control. In parallel, these technologies have leap-frogged drug discovery through identification of novel targets and drug specificity. Collectively, chemical proteomics technologies appear to do the impossible: uniting systematic analysis with a reductionist approach.
Collapse
Affiliation(s)
- Md Yousuf Ali
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Liron Bar-Peled
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
11
|
Bhargava A, Knapp JD, Fiehn O, Neylan TC, Inslicht SS. The lipidome of posttraumatic stress disorder. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.23.581833. [PMID: 38464224 PMCID: PMC10925102 DOI: 10.1101/2024.02.23.581833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Posttraumatic stress disorder (PTSD) can develop after trauma exposure. Some studies report that women develop PTSD at twice the rate of men, despite greater trauma exposure in men. Lipids and their metabolites (lipidome) regulate a myriad of key biological processes and pathways such as membrane integrity, oxidative stress, and neuroinflammation in the brain by maintaining neuronal connectivity and homeostasis. In this study, we analyzed the lipidome of 40 individuals with PTSD and 40 trauma-exposed non-PTSD individuals. Plasma samples were analyzed for lipidomics using Quadrupole Time-of-Flight (QToF) mass spectrometry. Additionally, ~ 90 measures were collected, on sleep, mental and physical health indices. Sleep quality worsened as PTSD severity increased in both sexes. The lipidomics analysis identified a total of 348 quantifiable known lipid metabolites and 1951 lipid metabolites that are yet unknown; known metabolites were part of 13 classes of lipids. After adjusting for sleep quality, in women with PTSD, only one lipid subclass, phosphatidylethanolamine (PE) was altered, whereas, in men with PTSD, 9 out of 13 subclasses were altered compared to non-PTSD women and men, respectively. Severe PTSD was associated with 22% and 5% of altered lipid metabolites in men and women, respectively. Of the changed metabolites, only 0.5% measures (2 PEs and cholesterol) were common between women and men with PTSD. Several sphingomyelins, PEs, ceramides, and triglycerides were increased in men with severe PTSD. The triglycerides and ceramide metabolites that were most highly increased were correlated with cholesterol metabolites and systolic blood pressure in men but not always in women with PTSD. Alterations in triglycerides and ceramides are linked with cardiac health and metabolic function in humans. Thus, disturbed sleep and higher weight may have contributed to changes in the lipidome found in PTSD.
Collapse
Affiliation(s)
- Aditi Bhargava
- Center for Reproductive Sciences, Department of Obstetrics and Gynecology, University of California San Francisco, CA 94143, USA
- Aseesa Inc., CA 94010, USA
| | | | - Oliver Fiehn
- NIH West Coast Metabolomics Center, University of California Davis Genome Center, Davis, CA 95616, USA
| | - Thomas C. Neylan
- San Francisco VA Health Care System, 4150 Clement St. (116P), San Francisco, CA 94121, USA
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, CA 94143, USA
| | - Sabra S. Inslicht
- San Francisco VA Health Care System, 4150 Clement St. (116P), San Francisco, CA 94121, USA
- Department of Psychiatry and Behavioral Sciences, University of California San Francisco, CA 94143, USA
| |
Collapse
|
12
|
Boseley RE, Sylvain NJ, Peeling L, Kelly ME, Pushie MJ. A review of concepts and methods for FTIR imaging of biomarker changes in the post-stroke brain. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2024; 1866:184287. [PMID: 38266967 DOI: 10.1016/j.bbamem.2024.184287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
Stroke represents a core area of study in neurosciences and public health due to its global contribution toward mortality and disability. The intricate pathophysiology of stroke, including ischemic and hemorrhagic events, involves the interruption in oxygen and nutrient delivery to the brain. Disruption of these crucial processes in the central nervous system leads to metabolic dysregulation and cell death. Fourier transform infrared (FTIR) spectroscopy can simultaneously measure total protein and lipid content along with a number of key biomarkers within brain tissue that cannot be observed using conventional techniques. FTIR imaging provides the opportunity to visualize this information in tissue which has not been chemically treated prior to analysis, thus retaining the spatial distribution and in situ chemical information. Here we present a review of FTIR imaging methods for investigating the biomarker responses in the post-stroke brain.
Collapse
Affiliation(s)
- Rhiannon E Boseley
- Department of Surgery, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
| | - Nicole J Sylvain
- Department of Surgery, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
| | - Lissa Peeling
- Department of Surgery, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
| | - Michael E Kelly
- Department of Surgery, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada
| | - M Jake Pushie
- Department of Surgery, University of Saskatchewan, Saskatoon, SK S7N 0W8, Canada.
| |
Collapse
|
13
|
Rostami-Nejad M, Asri N, Bakhtiari S, Khalkhal E, Maleki S, Rezaei-Tavirani M, Jahani-Sherafat S, Rostami K. Metabolomics and lipidomics signature in celiac disease: a narrative review. Clin Exp Med 2024; 24:34. [PMID: 38340186 PMCID: PMC10858823 DOI: 10.1007/s10238-024-01295-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 01/12/2024] [Indexed: 02/12/2024]
Abstract
Celiac disease (CD) is a chronic immune-mediated inflammatory disease of the small intestine caused by aberrant immune responses to consumed gluten proteins. CD is diagnosed by a combination of the patients reported symptoms, serologic and endoscopic biopsy evaluation of the small intestine; and adherence to a strict gluten-free diet (GFD) is considered the only available therapeutic approach for this disorder. Novel approaches need to be considered for finding new biomarkers to help this disorder diagnosis and finding a new alternative therapeutic method for this group of patients. Metabolomics and lipidomics are powerful tools to provide highly accurate and sensitive biomarkers. Previous studies indicated a metabolic fingerprint for CD deriving from alterations in gut microflora or intestinal permeability, malabsorption, and energy metabolism. Moreover, since CD is characterized by increased intestinal permeability and due to the importance of membrane lipid components in controlling barrier integrity, conducting lipidomics studies in this disorder is of great importance. In the current study, we tried to provide a critical overview of metabolomic and lipidomic changes in CD.
Collapse
Affiliation(s)
- Mohammad Rostami-Nejad
- Celiac Disease and Gluten Related Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Nastaran Asri
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sajjad Bakhtiari
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ensieh Khalkhal
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sepehr Maleki
- Department of Computer Science, University of Tabriz, Tabriz, Iran
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Somayeh Jahani-Sherafat
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kamran Rostami
- Department of Gastroenterology, MidCentral DHB, Palmerston North, 4442, New Zealand
| |
Collapse
|
14
|
Mondal S, Ghosh S. Liposome-Mediated Anti-Viral Drug Delivery Across Blood-Brain Barrier: Can Lipid Droplet Target Be Game Changers? Cell Mol Neurobiol 2023; 44:9. [PMID: 38123863 DOI: 10.1007/s10571-023-01443-4] [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: 07/18/2023] [Accepted: 12/02/2023] [Indexed: 12/23/2023]
Abstract
Lipid droplets (LDs) are subcellular organelles secreted from the endoplasmic reticulum (ER) that play a major role in lipid homeostasis. Recent research elucidates additional roles of LDs in cellular bioenergetics and innate immunity. LDs activate signaling cascades for interferon response and secretion of pro-inflammatory cytokines. Since balanced lipid homeostasis is critical for neuronal health, LDs play a crucial role in neurodegenerative diseases. RNA viruses enhance the secretion of LDs to support various phases of their life cycle in neurons which further leads to neurodegeneration. Targeting the excess LD formation in the brain could give us a new arsenal of antiviral therapeutics against neuroviruses. Liposomes are a suitable drug delivery system that could be used for drug delivery in the brain by crossing the Blood-Brain Barrier. Utilizing this, various pharmacological inhibitors and non-coding RNAs can be delivered that could inhibit the biogenesis of LDs or reduce their sizes, reversing the excess lipid-related imbalance in neurons. Liposome-Mediated Antiviral Drug Delivery Across Blood-Brain Barrier. Developing effective antiviral drug is challenging and it doubles against neuroviruses that needs delivery across the Blood-Brain Barrier (BBB). Lipid Droplets (LDs) are interesting targets for developing antivirals, hence targeting LD formation by drugs delivered using Liposomes can be game changers.
Collapse
Affiliation(s)
- Sourav Mondal
- CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India
| | - Sourish Ghosh
- CSIR-Indian Institute of Chemical Biology, 4 Raja S.C. Mullick Road, Jadavpur, Kolkata, West Bengal, 700032, India.
| |
Collapse
|
15
|
Solovyev N, Lucio M, Mandrioli J, Forcisi S, Kanawati B, Uhl J, Vinceti M, Schmitt-Kopplin P, Michalke B. Interplay of Metallome and Metabolome in Amyotrophic Lateral Sclerosis: A Study on Cerebrospinal Fluid of Patients Carrying Disease-Related Gene Mutations. ACS Chem Neurosci 2023; 14:3035-3046. [PMID: 37608584 PMCID: PMC10485893 DOI: 10.1021/acschemneuro.3c00128] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 08/09/2023] [Indexed: 08/24/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a lethal progressive neurodegenerative disease, characterized by a loss of function of upper and lower motor neurons. This study aimed to explore probable pathological alterations occurring in individuals with ALS compared to neurologically healthy controls through the analysis of cerebrospinal fluid (CSF), a medium, which directly interacts with brain parenchyma. A total of 7 ALS patients with disease-associated mutations (ATXN2, C9ORF72, FUS, SOD1, and TARDBP) and 13 controls were included in the study. Multiple analytical approaches were employed, including metabolomic and metallomics profiling, as well as genetic screening, using CSF samples obtained from the brain compartment. Data analysis involved the application of multivariate statistical methods. Advanced hyphenated selenium and redox metal (iron, copper, and manganese) speciation techniques and nontargeted Fourier transform ion cyclotron resonance mass spectrometry-based metabolomics were used for data acquisition. Nontargeted metabolomics showed reduced steroids, including sex hormones; additionally, copper and manganese species were found to be the most relevant features for ALS patients. This indicates a potential alteration of sex hormone pathways in the ALS-affected brain, as reflected in the CSF.
Collapse
Affiliation(s)
- Nikolay Solovyev
- Analytical
BioGeoChemistry Research Unit, Helmholtz
Center Munich—German Research Center for Environmental Health
GmbH, Ingolstädter
Landstr. 1, 85764 Neuherberg, Germany
| | - Marianna Lucio
- Analytical
BioGeoChemistry Research Unit, Helmholtz
Center Munich—German Research Center for Environmental Health
GmbH, Ingolstädter
Landstr. 1, 85764 Neuherberg, Germany
| | - Jessica Mandrioli
- Department
of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
- Department
of Neurosciences, Azienda Ospedaliero Universitaria
di Modena, 41126 Modena, Italy
| | - Sara Forcisi
- Analytical
BioGeoChemistry Research Unit, Helmholtz
Center Munich—German Research Center for Environmental Health
GmbH, Ingolstädter
Landstr. 1, 85764 Neuherberg, Germany
| | - Basem Kanawati
- Analytical
BioGeoChemistry Research Unit, Helmholtz
Center Munich—German Research Center for Environmental Health
GmbH, Ingolstädter
Landstr. 1, 85764 Neuherberg, Germany
| | - Jenny Uhl
- Analytical
BioGeoChemistry Research Unit, Helmholtz
Center Munich—German Research Center for Environmental Health
GmbH, Ingolstädter
Landstr. 1, 85764 Neuherberg, Germany
| | - Marco Vinceti
- CREAGEN
Research Center of Environmental, Genetic and Nutritional Epidemiology,
Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Philippe Schmitt-Kopplin
- Analytical
BioGeoChemistry Research Unit, Helmholtz
Center Munich—German Research Center for Environmental Health
GmbH, Ingolstädter
Landstr. 1, 85764 Neuherberg, Germany
| | - Bernhard Michalke
- Analytical
BioGeoChemistry Research Unit, Helmholtz
Center Munich—German Research Center for Environmental Health
GmbH, Ingolstädter
Landstr. 1, 85764 Neuherberg, Germany
| |
Collapse
|
16
|
Battis K, Xiang W, Winkler J. The Bidirectional Interplay of α-Synuclein with Lipids in the Central Nervous System and Its Implications for the Pathogenesis of Parkinson's Disease. Int J Mol Sci 2023; 24:13270. [PMID: 37686080 PMCID: PMC10487772 DOI: 10.3390/ijms241713270] [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: 07/31/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
The alteration and aggregation of alpha-synuclein (α-syn) play a crucial role in neurodegenerative diseases collectively termed as synucleinopathies, including Parkinson's disease (PD). The bidirectional interaction of α-syn with lipids and biomembranes impacts not only α-syn aggregation but also lipid homeostasis. Indeed, lipid composition and metabolism are severely perturbed in PD. One explanation for lipid-associated alterations may involve structural changes in α-syn, caused, for example, by missense mutations in the lipid-binding region of α-syn as well as post-translational modifications such as phosphorylation, acetylation, nitration, ubiquitination, truncation, glycosylation, and glycation. Notably, different strategies targeting the α-syn-lipid interaction have been identified and are able to reduce α-syn pathology. These approaches include the modulation of post-translational modifications aiming to reduce the aggregation of α-syn and modify its binding properties to lipid membranes. Furthermore, targeting enzymes involved in various steps of lipid metabolism and exploring the neuroprotective potential of lipids themselves have emerged as novel therapeutic approaches. Taken together, this review focuses on the bidirectional crosstalk of α-syn and lipids and how alterations of this interaction affect PD and thereby open a window for therapeutic interventions.
Collapse
Affiliation(s)
| | | | - Jürgen Winkler
- Department of Molecular Neurology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, 91054 Erlangen, Germany; (K.B.); (W.X.)
| |
Collapse
|
17
|
Maruyama T, Tanabe S, Uyeda A, Suzuki T, Muramatsu R. Free fatty acids support oligodendrocyte survival in a mouse model of amyotrophic lateral sclerosis. Front Cell Neurosci 2023; 17:1081190. [PMID: 37252191 PMCID: PMC10213402 DOI: 10.3389/fncel.2023.1081190] [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: 10/27/2022] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Introduction Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the white matter degeneration. Although changes in blood lipids are involved in the pathogenesis of neurological diseases, the pathological role of blood lipids in ALS remains unclear. Methods and results We performed lipidome analysis on the plasma of ALS model mice, mutant superoxide dismutase 1 (SOD1G93A) mice, and found that the concentration of free fatty acids (FFAs), including oleic acid (OA) and linoleic acid (LA), decreased prior to disease onset. An in vitro study revealed that OA and LA directly inhibited glutamate-induced oligodendrocytes cell death via free fatty acid receptor 1 (FFAR1). A cocktail containing OA/LA suppressed oligodendrocyte cell death in the spinal cord of SOD1G93A mice. Discussion These results suggested that the reduction of FFAs in the plasma is a pathogenic biomarker for ALS in the early stages, and supplying a deficiency in FFAs is a potential therapeutic approach for ALS by preventing oligodendrocyte cell death.
Collapse
Affiliation(s)
- Takashi Maruyama
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
- Department of Pharmacoscience, Graduate School of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Shogo Tanabe
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Akiko Uyeda
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Tatsunori Suzuki
- Department of Pharmacoscience, Graduate School of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
- Department of Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan
| | - Rieko Muramatsu
- Department of Molecular Pharmacology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
18
|
Janse van Mantgem MR, van Rheenen W, Hackeng AV, van Es MA, Veldink JH, van den Berg LH, van Eijk RPA. Association Between Serum Lipids and Survival in Patients With Amyotrophic Lateral Sclerosis: A Meta-analysis and Population-Based Study. Neurology 2023; 100:e1062-e1071. [PMID: 36460467 PMCID: PMC9990853 DOI: 10.1212/wnl.0000000000201657] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 10/20/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND AND OBJECTIVE To explore the association between lipids, polygenic profile scores (PPS) for biomarkers of lipid metabolism, markers of disease severity, and survival in patients with amyotrophic lateral sclerosis (ALS). METHODS We meta-analyzed the current literature on the prognostic value of lipids in patients with ALS. Subsequently, we evaluated the relationship between lipid levels at diagnosis, clinical disease stage, and survival in all consecutive patients diagnosed in the Netherlands. We determined the hazard ratio (HR) of each lipid for overall survival, defined as death from any cause. A subset of patients was matched to a previous genome-wide association study; data were used to calculate PPS for biomarkers of lipid metabolism and to determine the association between observed lipid levels at diagnosis and survival. RESULTS Meta-analysis of 4 studies indicated that none of the biomarkers of the lipid metabolism were statistically significantly associated with overall survival; there was, however, considerable heterogeneity between study results. Using individual patient data (N = 1,324), we found that increased high-density lipoprotein (HDL) cholesterol was associated with poorer survival (HR of 1.33 (95% CI 1.14-1.55, p < 0.001)). The correlation between BMI and HDL cholesterol (Pearson r -0.26, 95% CI -0.32 to -0.20) was negative and between BMI and triglycerides (TG) positive (Pearson r 0.18, 95% CI 0.12-0.24). Serum concentrations of total cholesterol and LDL cholesterol were lower in more advanced clinical stages (both p < 0.001). PPS for biomarkers of lipid metabolism explained 1.2%-13.1% of their variance at diagnosis. None of the PPS was significantly associated with survival (all p > 0.50). DISCUSSION Lipids may contain valuable information about disease severity and prognosis, but their main value may be driven as a consequence of disease progression. Our results underscore that gaining further insight into lipid metabolism and longitudinal data on serum concentrations of the lipid profile could improve the monitoring of patients and potentially further disentangle ALS pathogenesis.
Collapse
Affiliation(s)
- Mark R Janse van Mantgem
- From the Department of Neurology (M.R.J.M., W.R., A.V.H., M.A.E., J.H.V., L.H.B., R.P.A.E.), UMC Utrecht Brain Center, and Biostatistics & Research Support (R.P.A.E.), Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, The Netherlands
| | - Wouter van Rheenen
- From the Department of Neurology (M.R.J.M., W.R., A.V.H., M.A.E., J.H.V., L.H.B., R.P.A.E.), UMC Utrecht Brain Center, and Biostatistics & Research Support (R.P.A.E.), Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, The Netherlands
| | - Anemone V Hackeng
- From the Department of Neurology (M.R.J.M., W.R., A.V.H., M.A.E., J.H.V., L.H.B., R.P.A.E.), UMC Utrecht Brain Center, and Biostatistics & Research Support (R.P.A.E.), Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, The Netherlands
| | - Michael A van Es
- From the Department of Neurology (M.R.J.M., W.R., A.V.H., M.A.E., J.H.V., L.H.B., R.P.A.E.), UMC Utrecht Brain Center, and Biostatistics & Research Support (R.P.A.E.), Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, The Netherlands
| | - Jan H Veldink
- From the Department of Neurology (M.R.J.M., W.R., A.V.H., M.A.E., J.H.V., L.H.B., R.P.A.E.), UMC Utrecht Brain Center, and Biostatistics & Research Support (R.P.A.E.), Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, The Netherlands
| | - Leonard H van den Berg
- From the Department of Neurology (M.R.J.M., W.R., A.V.H., M.A.E., J.H.V., L.H.B., R.P.A.E.), UMC Utrecht Brain Center, and Biostatistics & Research Support (R.P.A.E.), Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, The Netherlands.
| | - Ruben P A van Eijk
- From the Department of Neurology (M.R.J.M., W.R., A.V.H., M.A.E., J.H.V., L.H.B., R.P.A.E.), UMC Utrecht Brain Center, and Biostatistics & Research Support (R.P.A.E.), Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, The Netherlands
| |
Collapse
|
19
|
Pfrieger FW. The Niemann-Pick type diseases – A synopsis of inborn errors in sphingolipid and cholesterol metabolism. Prog Lipid Res 2023; 90:101225. [PMID: 37003582 DOI: 10.1016/j.plipres.2023.101225] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/27/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
Disturbances of lipid homeostasis in cells provoke human diseases. The elucidation of the underlying mechanisms and the development of efficient therapies represent formidable challenges for biomedical research. Exemplary cases are two rare, autosomal recessive, and ultimately fatal lysosomal diseases historically named "Niemann-Pick" honoring the physicians, whose pioneering observations led to their discovery. Acid sphingomyelinase deficiency (ASMD) and Niemann-Pick type C disease (NPCD) are caused by specific variants of the sphingomyelin phosphodiesterase 1 (SMPD1) and NPC intracellular cholesterol transporter 1 (NPC1) or NPC intracellular cholesterol transporter 2 (NPC2) genes that perturb homeostasis of two key membrane components, sphingomyelin and cholesterol, respectively. Patients with severe forms of these diseases present visceral and neurologic symptoms and succumb to premature death. This synopsis traces the tortuous discovery of the Niemann-Pick diseases, highlights important advances with respect to genetic culprits and cellular mechanisms, and exposes efforts to improve diagnosis and to explore new therapeutic approaches.
Collapse
|
20
|
Xue M, Cao Y, Shen C, Guo W. Computational Advances of Protein/Neurotransmitter-membrane Interactions Involved in Vesicle Fusion and Neurotransmitter Release. J Mol Biol 2023; 435:167818. [PMID: 36089056 DOI: 10.1016/j.jmb.2022.167818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/22/2022] [Accepted: 09/04/2022] [Indexed: 02/04/2023]
Abstract
Vesicle fusion is of crucial importance to neuronal communication at neuron terminals. The exquisite but complex fusion machinery for neurotransmitter release is tightly controlled and regulated by protein/neurotransmitter-membrane interactions. Computational 'microscopies', in particular molecular dynamics simulations and related techniques, have provided notable insight into the physiological process over the past decades, and have made enormous contributions to fields such as neurology, pharmacology and pathophysiology. Here we review the computational advances of protein/neurotransmitter-membrane interactions related to presynaptic vesicle-membrane fusion and neurotransmitter release, and outline the in silico challenges ahead for understanding this important physiological process.
Collapse
Affiliation(s)
- Minmin Xue
- Key Laboratory for Intelligent Nano Materials and Devices of Ministry of Education, State Key Laboratory of Mechanics and Control of Mechanical Structures, and Institute for Frontier Science, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China
| | - Yuwei Cao
- State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, China
| | - Chun Shen
- Key Laboratory for Intelligent Nano Materials and Devices of Ministry of Education, State Key Laboratory of Mechanics and Control of Mechanical Structures, and Institute for Frontier Science, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China.
| | - Wanlin Guo
- Key Laboratory for Intelligent Nano Materials and Devices of Ministry of Education, State Key Laboratory of Mechanics and Control of Mechanical Structures, and Institute for Frontier Science, Nanjing University of Aeronautics and Astronautics, Nanjing 210016, China; State Key Laboratory of Tribology, Department of Mechanical Engineering, Tsinghua University, Beijing, China.
| |
Collapse
|
21
|
Agrawal I, Lim YS, Ng SY, Ling SC. Deciphering lipid dysregulation in ALS: from mechanisms to translational medicine. Transl Neurodegener 2022; 11:48. [DOI: 10.1186/s40035-022-00322-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/24/2022] [Indexed: 11/09/2022] Open
Abstract
AbstractLipids, defined by low solubility in water and high solubility in nonpolar solvents, can be classified into fatty acids, glycerolipids, glycerophospholipids, sphingolipids, and sterols. Lipids not only regulate integrity and fluidity of biological membranes, but also serve as energy storage and bioactive molecules for signaling. Causal mutations in SPTLC1 (serine palmitoyltransferase long chain subunit 1) gene within the lipogenic pathway have been identified in amyotrophic lateral sclerosis (ALS), a paralytic and fatal motor neuron disease. Furthermore, lipid dysmetabolism within the central nervous system and circulation is associated with ALS. Here, we aim to delineate the diverse roles of different lipid classes and understand how lipid dysmetabolism may contribute to ALS pathogenesis. Among the different lipids, accumulation of ceramides, arachidonic acid, and lysophosphatidylcholine is commonly emerging as detrimental to motor neurons. We end with exploring the potential ALS therapeutics by reducing these toxic lipids.
Collapse
|
22
|
Alterations in metabolic flux in migraine and the translational relevance. J Headache Pain 2022; 23:127. [PMID: 36175833 PMCID: PMC9523955 DOI: 10.1186/s10194-022-01494-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 09/10/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Migraine is a highly prevalent disorder with significant economical and personal burden. Despite the development of effective therapeutics, the causes which precipitate migraine attacks remain elusive. Clinical studies have highlighted altered metabolic flux and mitochondrial function in patients. In vivo animal experiments can allude to the metabolic mechanisms which may underlie migraine susceptibility. Understanding the translational relevance of these studies are important to identifying triggers, biomarkers and therapeutic targets in migraine. MAIN BODY Functional imaging studies have suggested that migraineurs feature metabolic syndrome, exhibiting hallmark features including upregulated oxidative phosphorylation yet depleted available free energy. Glucose hypometabolism is also evident in migraine patients and can lead to altered neuronal hyperexcitability such as the incidence of cortical spreading depression (CSD). The association between obesity and increased risk, frequency and worse prognosis of migraine also highlights lipid dysregulation in migraine pathology. Calcitonin gene related peptide (CGRP) has demonstrated an important role in sensitisation and nociception in headache, however its role in metabolic regulation in connection with migraine has not been thoroughly explored. Whether impaired metabolic function leads to increased release of peptides such as CGRP or excessive nociception leads to altered flux is yet unknown. CONCLUSION Migraine susceptibility may be underpinned by impaired metabolism resulting in depleted energy stores and altered neuronal function. This review discusses both clinical and in vivo studies which provide evidence of altered metabolic flux which contribute toward pathophysiology. It also reviews the translational relevance of animal studies in identifying targets of biomarker or therapeutic development.
Collapse
|
23
|
Lee TK, Lee JC, Kim D, Lee JW, Kim SS, Kim HI, Shin M, Cho J, Won MH, Choi S. Effects of Brain Factor‑7® against motor deficit and oxidative stress in a mouse model of MPTP‑induced Parkinson's disease. Exp Ther Med 2022; 24:635. [PMID: 36160902 PMCID: PMC9468851 DOI: 10.3892/etm.2022.11572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/01/2022] [Indexed: 11/17/2022] Open
Abstract
Oxidative stress is strongly implicated in the pathogenesis of Parkinson's disease (PD) through degeneration of dopaminergic neurons. The present study was designed to investigate the underlying mechanisms and therapeutic potential of Brain Factor-7® (BF-7®), a natural compound in silkworm, in a mouse model of PD induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). MPTP (20 mg/kg) was intraperitoneally injected into mice to cause symptoms of PD. Mice were orally administered BF-7® (a mixture of silk peptides) before and after MPTP treatment. Rotarod performance test was used to assess motor performance. Fluoro-Jade B staining for neurons undergoing degeneration and immunohistochemistry of tyrosine hydroxylase for dopaminergic neurons, 4-hydroxy-2-nonenal (4HNE) for lipid peroxidation, 8-hydroxy-2'-deoxyguanosine (8OHdG) for DNA damage and superoxide dismutase (SOD) 1 and SOD2 for antioxidative enzymes in the pars compacta of the substantia nigra were performed. Results showed that BF-7® treatment significantly improved MPTP-induced motor deficit and protected MPTP-induced dopaminergic neurodegeneration. Furthermore, BF-7® treatment significantly ameliorated MPTP-induced oxidative stress. Increased 4HNE and 8OHdG immunoreactivities induced by MPTP were significantly reduced by BF-7®, whereas SOD1 and SOD2 immunoreactivities decreased by MPTP were significantly enhanced by BF-7®. In conclusion, BF-7® exerted protective and/or therapeutic effects in a mouse model of PD by decreasing effects of oxidative stress on dopaminergic neurons in the substantia nigra pars compacta.
Collapse
Affiliation(s)
- Tae-Kyeong Lee
- Department of Food Science and Nutrition, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| | - Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Dae Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangnung‑Wonju National University, Gangneung 25457, Republic of Korea
| | - Ji-Won Lee
- Precision Medicine R&D Center, Famenity Co., Ltd., Uiwang, Gyeonggi 16006, Republic of Korea
| | - Sung-Su Kim
- Precision Medicine R&D Center, Famenity Co., Ltd., Uiwang, Gyeonggi 16006, Republic of Korea
| | - Hyung-Il Kim
- Department of Emergency Medicine, Dankook University Hospital, College of Medicine, Dankook University, Cheonan, Chungnam 31116, Republic of Korea
| | - Myoung Shin
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24289, Republic of Korea
| | - Jun Cho
- Department of Emergency Medicine, Kangwon National University Hospital, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24289, Republic of Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Soo Choi
- Department of Biomedical Science, Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, Gangwon 24252, Republic of Korea
| |
Collapse
|
24
|
Lanznaster D, Bruno C, Bourgeais J, Emond P, Zemmoura I, Lefèvre A, Reynier P, Eymieux S, Blanchard E, Vourc’h P, Andres CR, Bakkouche SE, Herault O, Favard L, Corcia P, Blasco H. Metabolic Profile and Pathological Alterations in the Muscle of Patients with Early-Stage Amyotrophic Lateral Sclerosis. Biomedicines 2022; 10:biomedicines10061307. [PMID: 35740329 PMCID: PMC9220134 DOI: 10.3390/biomedicines10061307] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/30/2022] [Accepted: 05/30/2022] [Indexed: 12/26/2022] Open
Abstract
Diverse biomarkers and pathological alterations have been found in muscle of patients with Amyotrophic lateral sclerosis (ALS), but the relation between such alterations and dysfunction in energetic metabolism remains to be investigated. We established the metabolome of muscle and serum of ALS patients and correlated these findings with the clinical status and pathological alterations observed in the muscle. We obtained data from 20 controls and 17 ALS patients (disease duration: 9.4 ± 6.8 months). Multivariate metabolomics analysis identified a distinct serum metabolome for ALS compared to controls (p-CV-ANOVA < 0.035) and revealed an excellent discriminant profile for muscle metabolome (p-CV-ANOVA < 0.0012). Citramalate was discriminant for both muscle and serum. High lauroylcarnitine levels in muscle were associated with low Forced Vital Capacity. Transcriptomics analysis of key antioxidant enzymes showed an upregulation of SOD3 (p = 0.0017) and GLRX2(1) (p = 0.0022) in ALS muscle. Analysis of mitochondrial enzymatic activity in muscle revealed higher complex II/CS (p = 0.04) and lower LDH (p = 0.03) activity in ALS than in controls. Our study showed, for the first time, a global dysfunction in the muscle of early-stage ALS patients. Furthermore, we identified novel metabolites to be employed as biomarkers for diagnosis and prognosis of ALS patients.
Collapse
Affiliation(s)
- Débora Lanznaster
- UMR 1253, iBrain, Université de Tours, INSERM, 37000 Tours, France; (C.B.); (P.E.); (I.Z.); (A.L.); (P.V.); (C.R.A.); (P.C.); (H.B.)
- Correspondence:
| | - Clément Bruno
- UMR 1253, iBrain, Université de Tours, INSERM, 37000 Tours, France; (C.B.); (P.E.); (I.Z.); (A.L.); (P.V.); (C.R.A.); (P.C.); (H.B.)
- Service de Biochimie et Biologie Moléculaire, CHU de Tours, 37000 Tours, France
| | - Jérôme Bourgeais
- CNRS ERL7001, EA 7501 GICC, Université de Tours, 37000 Tours, France; (J.B.); (O.H.)
| | - Patrick Emond
- UMR 1253, iBrain, Université de Tours, INSERM, 37000 Tours, France; (C.B.); (P.E.); (I.Z.); (A.L.); (P.V.); (C.R.A.); (P.C.); (H.B.)
- Service de Médecine Nucléaire In Vitro, CHU de Tours, 37000 Tours, France
| | - Ilyess Zemmoura
- UMR 1253, iBrain, Université de Tours, INSERM, 37000 Tours, France; (C.B.); (P.E.); (I.Z.); (A.L.); (P.V.); (C.R.A.); (P.C.); (H.B.)
- Service de Neurochirurgie, CHU de Tours, 37000 Tours, France
| | - Antoine Lefèvre
- UMR 1253, iBrain, Université de Tours, INSERM, 37000 Tours, France; (C.B.); (P.E.); (I.Z.); (A.L.); (P.V.); (C.R.A.); (P.C.); (H.B.)
| | - Pascal Reynier
- Service de Biochimie et Biologie Moléculaire, CHU d’Angers, 49000 Angers, France;
- Mitovasc-Mitolab, UMR CNRS6015-INSERM1083, 49000 Angers, France
| | - Sébastien Eymieux
- Plateforme IBiSA de Microscopie Electronique, Université de Tours et CHU de Tours, 37000 Tours, France; (S.E.); (E.B.)
- INSERM U1259, Université de Tours, 37000 Tours, France
| | - Emmanuelle Blanchard
- Plateforme IBiSA de Microscopie Electronique, Université de Tours et CHU de Tours, 37000 Tours, France; (S.E.); (E.B.)
- INSERM U1259, Université de Tours, 37000 Tours, France
| | - Patrick Vourc’h
- UMR 1253, iBrain, Université de Tours, INSERM, 37000 Tours, France; (C.B.); (P.E.); (I.Z.); (A.L.); (P.V.); (C.R.A.); (P.C.); (H.B.)
- Service de Biochimie et Biologie Moléculaire, CHU de Tours, 37000 Tours, France
| | - Christian R. Andres
- UMR 1253, iBrain, Université de Tours, INSERM, 37000 Tours, France; (C.B.); (P.E.); (I.Z.); (A.L.); (P.V.); (C.R.A.); (P.C.); (H.B.)
- Service de Biochimie et Biologie Moléculaire, CHU de Tours, 37000 Tours, France
| | | | - Olivier Herault
- CNRS ERL7001, EA 7501 GICC, Université de Tours, 37000 Tours, France; (J.B.); (O.H.)
| | - Luc Favard
- Service de Neurologie, CHU de Tours, 37000 Tours, France;
| | - Philippe Corcia
- UMR 1253, iBrain, Université de Tours, INSERM, 37000 Tours, France; (C.B.); (P.E.); (I.Z.); (A.L.); (P.V.); (C.R.A.); (P.C.); (H.B.)
- Service de Neurologie, CHU de Tours, 37000 Tours, France;
| | - Hélène Blasco
- UMR 1253, iBrain, Université de Tours, INSERM, 37000 Tours, France; (C.B.); (P.E.); (I.Z.); (A.L.); (P.V.); (C.R.A.); (P.C.); (H.B.)
- Service de Biochimie et Biologie Moléculaire, CHU de Tours, 37000 Tours, France
| |
Collapse
|
25
|
Godoy-Corchuelo JM, Fernández-Beltrán LC, Ali Z, Gil-Moreno MJ, López-Carbonero JI, Guerrero-Sola A, Larrad-Sainz A, Matias-Guiu J, Matias-Guiu JA, Cunningham TJ, Corrochano S. Lipid Metabolic Alterations in the ALS-FTD Spectrum of Disorders. Biomedicines 2022; 10:1105. [PMID: 35625841 PMCID: PMC9138405 DOI: 10.3390/biomedicines10051105] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 02/06/2023] Open
Abstract
There is an increasing interest in the study of the relation between alterations in systemic lipid metabolism and neurodegenerative disorders, in particular in Amyotrophic Lateral Sclerosis (ALS) and Frontotemporal Dementia (FTD). In ALS these alterations are well described and evident not only with the progression of the disease but also years before diagnosis. Still, there are some discrepancies in findings relating to the causal nature of lipid metabolic alterations, partly due to the great clinical heterogeneity in ALS. ALS presentation is within a disorder spectrum with Frontotemporal Dementia (FTD), and many patients present mixed forms of ALS and FTD, thus increasing the variability. Lipid metabolic and other systemic metabolic alterations have not been well studied in FTD, or in ALS-FTD mixed forms, as has been in pure ALS. With the recent development in lipidomics and the integration with other -omics platforms, there is now emerging data that not only facilitates the identification of biomarkers but also enables understanding of the underlying pathological mechanisms. Here, we reviewed the recent literature to compile lipid metabolic alterations in ALS, FTD, and intermediate mixed forms, with a view to appraising key commonalities or differences within the spectrum.
Collapse
Affiliation(s)
- Juan Miguel Godoy-Corchuelo
- Neurological Disorders Group, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain; (J.M.G.-C.); (L.C.F.-B.); (M.J.G.-M.); (J.I.L.-C.); (A.G.-S.); (J.M.-G.); (J.A.M.-G.)
| | - Luis C. Fernández-Beltrán
- Neurological Disorders Group, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain; (J.M.G.-C.); (L.C.F.-B.); (M.J.G.-M.); (J.I.L.-C.); (A.G.-S.); (J.M.-G.); (J.A.M.-G.)
| | - Zeinab Ali
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (Z.A.); (T.J.C.)
| | - María J. Gil-Moreno
- Neurological Disorders Group, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain; (J.M.G.-C.); (L.C.F.-B.); (M.J.G.-M.); (J.I.L.-C.); (A.G.-S.); (J.M.-G.); (J.A.M.-G.)
| | - Juan I. López-Carbonero
- Neurological Disorders Group, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain; (J.M.G.-C.); (L.C.F.-B.); (M.J.G.-M.); (J.I.L.-C.); (A.G.-S.); (J.M.-G.); (J.A.M.-G.)
| | - Antonio Guerrero-Sola
- Neurological Disorders Group, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain; (J.M.G.-C.); (L.C.F.-B.); (M.J.G.-M.); (J.I.L.-C.); (A.G.-S.); (J.M.-G.); (J.A.M.-G.)
| | - Angélica Larrad-Sainz
- Nutrition and Endocrinology Group, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain;
| | - Jorge Matias-Guiu
- Neurological Disorders Group, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain; (J.M.G.-C.); (L.C.F.-B.); (M.J.G.-M.); (J.I.L.-C.); (A.G.-S.); (J.M.-G.); (J.A.M.-G.)
| | - Jordi A. Matias-Guiu
- Neurological Disorders Group, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain; (J.M.G.-C.); (L.C.F.-B.); (M.J.G.-M.); (J.I.L.-C.); (A.G.-S.); (J.M.-G.); (J.A.M.-G.)
| | - Thomas J. Cunningham
- MRC Harwell Institute, Harwell Campus, Oxfordshire OX11 0RD, UK; (Z.A.); (T.J.C.)
- MRC Prion Unit at UCL, UCL Institute of Prion Diseases, London W1W 7FF, UK
| | - Silvia Corrochano
- Neurological Disorders Group, Hospital Clínico San Carlos, IdISSC, 28040 Madrid, Spain; (J.M.G.-C.); (L.C.F.-B.); (M.J.G.-M.); (J.I.L.-C.); (A.G.-S.); (J.M.-G.); (J.A.M.-G.)
| |
Collapse
|
26
|
Hop PJ, Zwamborn RA, Hannon E, Shireby GL, Nabais MF, Walker EM, van Rheenen W, van Vugt JJ, Dekker AM, Westeneng HJ, Tazelaar GH, van Eijk KR, Moisse M, Baird D, Khleifat AA, Iacoangeli A, Ticozzi N, Ratti A, Cooper-Knock J, Morrison KE, Shaw PJ, Basak AN, Chiò A, Calvo A, Moglia C, Canosa A, Brunetti M, Grassano M, Gotkine M, Lerner Y, Zabari M, Vourc’h P, Corcia P, Couratier P, Pardina JSM, Salas T, Dion P, Ross JP, Henderson RD, Mathers S, McCombe PA, Needham M, Nicholson G, Rowe DB, Pamphlett R, Mather KA, Sachdev PS, Furlong S, Garton FC, Henders AK, Lin T, Ngo ST, Steyn FJ, Wallace L, Williams KL, Neto MM, Cauchi RJ, Blair IP, Kiernan MC, Drory V, Povedano M, de Carvalho M, Pinto S, Weber M, Rouleau GA, Silani V, Landers JE, Shaw CE, Andersen PM, McRae AF, van Es MA, Pasterkamp RJ, Wray NR, McLaughlin RL, Hardiman O, Kenna KP, Tsai E, Runz H, Al-Chalabi A, van den Berg LH, Van Damme P, Mill J, Veldink JH. Genome-wide study of DNA methylation shows alterations in metabolic, inflammatory, and cholesterol pathways in ALS. Sci Transl Med 2022; 14:eabj0264. [PMID: 35196023 PMCID: PMC10040186 DOI: 10.1126/scitranslmed.abj0264] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease with an estimated heritability between 40 and 50%. DNA methylation patterns can serve as proxies of (past) exposures and disease progression, as well as providing a potential mechanism that mediates genetic or environmental risk. Here, we present a blood-based epigenome-wide association study meta-analysis in 9706 samples passing stringent quality control (6763 patients, 2943 controls). We identified a total of 45 differentially methylated positions (DMPs) annotated to 42 genes, which are enriched for pathways and traits related to metabolism, cholesterol biosynthesis, and immunity. We then tested 39 DNA methylation-based proxies of putative ALS risk factors and found that high-density lipoprotein cholesterol, body mass index, white blood cell proportions, and alcohol intake were independently associated with ALS. Integration of these results with our latest genome-wide association study showed that cholesterol biosynthesis was potentially causally related to ALS. Last, DNA methylation at several DMPs and blood cell proportion estimates derived from DNA methylation data were associated with survival rate in patients, suggesting that they might represent indicators of underlying disease processes potentially amenable to therapeutic interventions.
Collapse
Affiliation(s)
- Paul J. Hop
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - Ramona A.J. Zwamborn
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - Eilis Hannon
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter EX1 2LU, UK
| | - Gemma L. Shireby
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter EX1 2LU, UK
| | - Marta F. Nabais
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter EX1 2LU, UK
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| | - Emma M. Walker
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter EX1 2LU, UK
| | - Wouter van Rheenen
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - Joke J.F.A. van Vugt
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - Annelot M. Dekker
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - Henk-Jan Westeneng
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - Gijs H.P. Tazelaar
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - Kristel R. van Eijk
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - Matthieu Moisse
- KU Leuven–University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), Leuven 3000, Belgium
- VIB, Center for Brain and Disease Research, Leuven 3000, Belgium
- University Hospitals Leuven, Department of Neurology, Leuven 3000, Belgium
| | - Denis Baird
- Translational Biology, Biogen, Boston, MA 02142, USA
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, University of Bristol, Bristol BS8 2BN, UK
| | - Ahmad Al Khleifat
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
| | - Alfredo Iacoangeli
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
- National Institute for Health Research Biomedical Research Centre and Dementia Unit, South London and Maudsley NHS Foundation Trust and King’s College London, London SE5 8AZ, UK
| | - Nicola Ticozzi
- Department of Neurology-Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan 20149, Italy
- Department of Pathophysiology and Transplantation, “Dino Ferrari” Center, Università degli Studi di Milano, Milan 20122, Italy
| | - Antonia Ratti
- Department of Neurology-Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan 20149, Italy
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milano 20145, Italy
| | - Jonathan Cooper-Knock
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield S10 2HQ, UK
| | - Karen E. Morrison
- School of Medicine, Dentistry, and Biomedical Sciences, Queen’s University Belfast, Belfast BT9 7BL, UK
| | - Pamela J. Shaw
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield S10 2HQ, UK
| | - A. Nazli Basak
- Koc University, School of Medicine, Translational Medicine Research Center, NDAL, Istanbul, 34450, Turkey
| | - Adriano Chiò
- “Rita Levi Montalcini” Department of Neuroscience, ALS Centre, University of Torino, Turin 10126, Italy
- Azienda Ospedaliero-Universitaria Città della Salute e della Scienza, SC Neurologia 1U, Turin 10126, Italy
| | - Andrea Calvo
- “Rita Levi Montalcini” Department of Neuroscience, ALS Centre, University of Torino, Turin 10126, Italy
- Azienda Ospedaliero-Universitaria Città della Salute e della Scienza, SC Neurologia 1U, Turin 10126, Italy
| | - Cristina Moglia
- “Rita Levi Montalcini” Department of Neuroscience, ALS Centre, University of Torino, Turin 10126, Italy
- Azienda Ospedaliero-Universitaria Città della Salute e della Scienza, SC Neurologia 1U, Turin 10126, Italy
| | - Antonio Canosa
- “Rita Levi Montalcini” Department of Neuroscience, ALS Centre, University of Torino, Turin 10126, Italy
- Azienda Ospedaliero-Universitaria Città della Salute e della Scienza, SC Neurologia 1U, Turin 10126, Italy
| | - Maura Brunetti
- “Rita Levi Montalcini” Department of Neuroscience, ALS Centre, University of Torino, Turin 10126, Italy
| | - Maurizio Grassano
- “Rita Levi Montalcini” Department of Neuroscience, ALS Centre, University of Torino, Turin 10126, Italy
| | - Marc Gotkine
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91904, Israel
- Agnes Ginges Center for Human Neurogenetics, Department of Neurology, Hadassah Medical Center, Jerusalem 91120, Israel
| | - Yossef Lerner
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91904, Israel
- Agnes Ginges Center for Human Neurogenetics, Department of Neurology, Hadassah Medical Center, Jerusalem 91120, Israel
| | - Michal Zabari
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 91904, Israel
- Agnes Ginges Center for Human Neurogenetics, Department of Neurology, Hadassah Medical Center, Jerusalem 91120, Israel
| | - Patrick Vourc’h
- Service de Biochimie et Biologie moléculaire, CHU de Tours, Tours 37044, France
- UMR 1253, Université de Tours, Inserm, Tours 37044, France
| | - Philippe Corcia
- UMR 1253, Université de Tours, Inserm, Tours 37044, France
- Centre de référence sur la SLA, CHU de Tours, Tours 37044, France
| | - Philippe Couratier
- Centre de référence sur la SLA, CHRU de Limoges, Limoges 87042, France
- UMR 1094, Université de Limoges, Inserm, Limoges 87025, France
| | | | - Teresa Salas
- Department of Neurology, Hospital La Paz-Carlos III, Madrid 28046, Spain
| | - Patrick Dion
- Montréal Neurological Institute and Hospital, McGill University, Montréal, QC H3A 2B4, Canada
| | - Jay P. Ross
- Montréal Neurological Institute and Hospital, McGill University, Montréal, QC H3A 2B4, Canada
- Department of Human Genetics, McGill University, Montréal, QC H3A 0C7, Canada
| | - Robert D. Henderson
- Department of Neurology, Royal Brisbane and Women’s Hospital, Brisbane, QLD 4029, Australia
| | - Susan Mathers
- Calvary Health Care Bethlehem, Parkdale, VIC 3195, Australia
| | - Pamela A. McCombe
- Centre for Clinical Research, University of Queensland, Brisbane, QLD 4019, Australia
| | - Merrilee Needham
- Fiona Stanley Hospital, Perth, WA 6150, Australia
- Notre Dame University, Fremantle, WA 6160, Australia
- Institute for Immunology and Infectious Diseases, Murdoch University, Perth, WA 6150, Australia
| | - Garth Nicholson
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney, NSW 2139, Australia
| | - Dominic B. Rowe
- Centre for Motor Neuron Disease Research, Macquarie University, NSW 2109, Australia
| | - Roger Pamphlett
- Discipline of Pathology and Department of Neuropathology, Brain and Mind Centre, University of Sydney, Sydney, NSW 2050, Australia
| | - Karen A. Mather
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW 2031, Australia
- Neuroscience Research Australia Institute, Randwick, NSW 2031, Australia
| | - Perminder S. Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, University of New South Wales, Sydney, NSW 2031, Australia
- Neuropsychiatric Institute, Prince of Wales Hospital, UNSW, Randwick, NSW 2031, Australia
| | - Sarah Furlong
- Centre for Motor Neuron Disease Research, Macquarie University, NSW 2109, Australia
| | - Fleur C. Garton
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| | - Anjali K. Henders
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| | - Tian Lin
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| | - Shyuan T. Ngo
- Centre for Clinical Research, University of Queensland, Brisbane, QLD 4019, Australia
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, QLD 4072, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia
| | - Frederik J. Steyn
- Centre for Clinical Research, University of Queensland, Brisbane, QLD 4019, Australia
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD 4072, Australia
| | - Leanne Wallace
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| | - Kelly L. Williams
- Centre for Motor Neuron Disease Research, Macquarie University, NSW 2109, Australia
| | | | | | | | - Ruben J. Cauchi
- Center for Molecular Medicine and Biobanking and Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, 2023 Msida, Malta
| | - Ian P. Blair
- Centre for Motor Neuron Disease Research, Macquarie University, NSW 2109, Australia
| | - Matthew C. Kiernan
- Brain and Mind Centre, University of Sydney, Sydney, NSW, 2050, Australia
- Department of Neurology, Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
| | - Vivian Drory
- Department of Neurology, Tel-Aviv Sourasky Medical Centre, Tel-Aviv 64239, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Monica Povedano
- Functional Unit of Amyotrophic Lateral Sclerosis (UFELA), Service of Neurology, Bellvitge University Hospital, L’Hospitalet de Llobregat, Barcelona 08907, Spain
| | - Mamede de Carvalho
- Instituto de Fisiologia, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon 1649-028, Portugal
| | - Susana Pinto
- Instituto de Fisiologia, Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon 1649-028, Portugal
| | - Markus Weber
- Neuromuscular Diseases Unit/ALS Clinic, Kantonsspital St. Gallen, 9007 St. Gallen, Switzerland
| | - Guy A. Rouleau
- Montréal Neurological Institute and Hospital, McGill University, Montréal, QC H3A 2B4, Canada
| | - Vincenzo Silani
- Department of Neurology-Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano IRCCS, Milan 20149, Italy
- Department of Pathophysiology and Transplantation, “Dino Ferrari” Center, Università degli Studi di Milano, Milan 20122, Italy
| | - John E. Landers
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Christopher E. Shaw
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
| | - Peter M. Andersen
- Department of Clinical Science, Umeå University, Umeå SE-901 85, Sweden
| | - Allan F. McRae
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
| | - Michael A. van Es
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - R. Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, 3584 CX, Netherlands
| | - Naomi R. Wray
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD4072, Australia
- Queensland Brain Institute, University of Queensland, Brisbane, QLD 4072, Australia
| | - Russell L. McLaughlin
- Complex Trait Genomics Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, Dublin D02 PN40, Ireland
| | - Orla Hardiman
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin D02 PN40, Ireland
| | - Kevin P. Kenna
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, 3584 CX, Netherlands
| | - Ellen Tsai
- Translational Biology, Biogen, Boston, MA 02142, USA
| | - Heiko Runz
- Translational Biology, Biogen, Boston, MA 02142, USA
| | - Ammar Al-Chalabi
- Maurice Wohl Clinical Neuroscience Institute, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London SE5 8AF, UK
- King’s College Hospital, Denmark Hill, London SE5 9RS, UK
| | - Leonard H. van den Berg
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| | - Philip Van Damme
- KU Leuven–University of Leuven, Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), Leuven 3000, Belgium
- VIB, Center for Brain and Disease Research, Leuven 3000, Belgium
- University Hospitals Leuven, Department of Neurology, Leuven 3000, Belgium
| | - Jonathan Mill
- University of Exeter Medical School, College of Medicine and Health, University of Exeter, Exeter EX1 2LU, UK
| | - Jan H. Veldink
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht 3584 CX, Netherlands
| |
Collapse
|
27
|
Qin Q, Yin Y, Xing Y, Wang X, Wang Y, Wang F, Tang Y. Lipid Metabolism in the Development and Progression of Vascular Cognitive Impairment: A Systematic Review. Front Neurol 2021; 12:709134. [PMID: 34867708 PMCID: PMC8639494 DOI: 10.3389/fneur.2021.709134] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 10/25/2021] [Indexed: 11/27/2022] Open
Abstract
Background: Vascular cognitive impairment (VCI) is a major public health problem. The current diagnosis of VCI is made based on the assessment of clinical symptoms and neuropsychological measurements, and is supported by neuroimaging. These methods are both time-consuming and expensive, which leads to needs for alternative biomarkers for VCI. Metabolomics is an emerging and powerful tool to discover of new biomarkers of disease, which can investigate variations in different metabolic processes such as lipid, since the brain is highly enriched in lipids and that lipid changes may lead to pathology in the brain. Vascular cognitive impairment is vulnerable to the disturbance of lipid metabolism. Furthermore, blood samples, which could be identified as reliable clinical biomarkers are relatively convenient to obtain and provide a non-invasive assessment. Therefore, our study aims to understand whether peripheral lipid biomarkers can be used as diagnostic biomarkers and monitor the progression of VCI. Methods: We systematically searched the PubMed, Embase, CNKI, and VIP databases to find VCI and lipid metabolism in reports from inception through February 2021. Studies meeting the following criteria were eligible: (1) original studies in humans; (2) lipid metabolites in blood; (3) reports of VCI. Results: Through our review, nine original articles were eligible. Blood-based metabolites that might be potential biomarkers were identified. Most of them including PC, PE, Cers, and ChEs were significantly lower, while elevation of FAs and DGs were associated with VCI. Most importantly, these blood-based metabolites might be proposed as potential biomarkers for VCI, which provides direction for further validation. Discussion and Conclusion: To the best of our knowledge, this is the first systemic review concerning the relationship of lipid metabolism and VCI. It identifies potential biomarkers and provides insights into the disease pathobiology. However, more advanced studies and researches on a lipidomic platform must be done to understand the exact pathology behind and identify potential lipid biomarkers, which might help achieve the goal of discovering novel therapeutics.
Collapse
Affiliation(s)
- Qi Qin
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Yunsi Yin
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Yi Xing
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Xuan Wang
- Department of Endocrinology, Mudanjiang Second People's Hospital, Mudanjiang, China
| | - Yan Wang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Fan Wang
- Department of Epidemiology, School of Public Health, Harbin Medical University, Harbin, China
- *Correspondence: Fan Wang
| | - Yi Tang
- Innovation Center for Neurological Disorders, Department of Neurology, Xuanwu Hospital, Capital Medical University, National Clinical Research Center for Geriatric Diseases, Beijing, China
- Neurodegenerative Laboratory of Ministry of Education of the People's Republic of China, Beijing Key Laboratory of Geriatric Cognitive Disorders, Beijing, China
- Yi Tang
| |
Collapse
|
28
|
Cassereau J, Corcia P, Reynier P. Towards personalized medicine for amyotrophic lateral sclerosis. Trends Endocrinol Metab 2021; 32:839-841. [PMID: 34325980 DOI: 10.1016/j.tem.2021.07.002] [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: 06/21/2021] [Accepted: 07/09/2021] [Indexed: 10/20/2022]
Abstract
Mohassel et al. provide unprecedented dichotomy of consequences on sphingolipid biosynthesis between pathogenic variants in the SPTLC1 gene, responsible for either amyotrophic lateral sclerosis (ALS) or hereditary sensory and autonomic neuropathy type 1 (HSAN1). Normalization of sphingolipid levels by siRNA selectively targeting the ALS mutant allele mRNA sheds light on new therapeutic approaches.
Collapse
Affiliation(s)
- Julien Cassereau
- Department of Neurology, University Hospital, Angers, France; Amyotrophic Lateral Sclerosis (ALS) Center, University Hospital, Angers, France; University of Angers, Institut National de la Santé et de la Recherche Médicale (INSERM) and Centre National de la Recherche Scientifique (CNRS) Mitochondrial and Cardiovascular Physiopathology (MitoVasc) Institute, Angers, France.
| | - Philippe Corcia
- ALS Center, Centre Hospitalier Universitaire (CHU) Bretonneau, Tours, France; INSERM Unit U1253 Imaging and Brain, Tours, France
| | - Pascal Reynier
- University of Angers, Institut National de la Santé et de la Recherche Médicale (INSERM) and Centre National de la Recherche Scientifique (CNRS) Mitochondrial and Cardiovascular Physiopathology (MitoVasc) Institute, Angers, France; Department of Biochemistry and Molecular Biology, University Hospital, Angers, France
| |
Collapse
|
29
|
Histone Deacetylase Inhibition Regulates Lipid Homeostasis in a Mouse Model of Amyotrophic Lateral Sclerosis. Int J Mol Sci 2021; 22:ijms222011224. [PMID: 34681883 PMCID: PMC8541517 DOI: 10.3390/ijms222011224] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/08/2021] [Accepted: 10/15/2021] [Indexed: 12/19/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an incurable and fatal neurodegenerative disorder of the motor system. While the etiology is still incompletely understood, defects in metabolism act as a major contributor to the disease progression. Recently, histone deacetylase (HDAC) inhibition using ACY-738 has been shown to restore metabolic alterations in the spinal cord of a FUS mouse model of ALS, which was accompanied by a beneficial effect on the motor phenotype and survival. In this study, we investigated the specific effects of HDAC inhibition on lipid metabolism using untargeted lipidomic analysis combined with transcriptomic analysis in the spinal cord of FUS mice. We discovered that symptomatic FUS mice recapitulate lipid alterations found in ALS patients and in the SOD1 mouse model. Glycerophospholipids, sphingolipids, and cholesterol esters were most affected. Strikingly, HDAC inhibition mitigated lipid homeostasis defects by selectively targeting glycerophospholipid metabolism and reducing cholesteryl esters accumulation. Therefore, our data suggest that HDAC inhibition is a potential new therapeutic strategy to modulate lipid metabolism defects in ALS and potentially other neurodegenerative diseases.
Collapse
|
30
|
Galkina OV, Vetrovoy OV, Eschenko ND. The Role of Lipids in Implementing Specific Functions in the Central Nervous System. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2021. [DOI: 10.1134/s1068162021050253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
31
|
Li X, Pu Y, Xu Y, Cao J, Jiang W. Potential Hypolipidemic Effects of Banana Condensed Tannins Through the Interaction with Digestive Juice Components Related to Lipid Digestion. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:8703-8713. [PMID: 34324317 DOI: 10.1021/acs.jafc.1c02794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
An in vitro intestinal model was used to evaluate the impact of banana condensed tannins (BCT) on the digestion of lipids (fat and cholesterol). BCT significantly suppressed the digestion of fat and cholesterol by interacting with digestive juice components. The interactions of BCT with a digestive juice mixture and its components (including bile acid, lipase, cholesterol esterase, CaCl2, NaCl, and cholesterol) were analyzed using turbidity, isothermal titration calorimetry, particle size distribution, zeta potential, and molecular docking analyses. The results showed that BCT reduced the digestion of lipids mainly via interaction with lipase, cholesterol esterase, bile acid, and cholesterol. Electrostatic CT-calcium ion complexes might reduce the extent of lipid digestion by decreasing the surface area of the lipid droplets exposed to the enzymes. This research provides valuable insights into the molecular mechanisms of the interaction of BCT with digestive juice components related to lipid digestion that may affect the rate and extent of lipid digestion.
Collapse
Affiliation(s)
- Xiangxin Li
- College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghuadonglu Road, Beijing 100083, P. R. China
| | - Yijing Pu
- College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghuadonglu Road, Beijing 100083, P. R. China
| | - Yan Xu
- College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghuadonglu Road, Beijing 100083, P. R. China
| | - Jiankang Cao
- College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghuadonglu Road, Beijing 100083, P. R. China
| | - Weibo Jiang
- College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghuadonglu Road, Beijing 100083, P. R. China
| |
Collapse
|
32
|
Stella R, Bonadio RS, Cagnin S, Massimino ML, Bertoli A, Peggion C. Perturbations of the Proteome and of Secreted Metabolites in Primary Astrocytes from the hSOD1(G93A) ALS Mouse Model. Int J Mol Sci 2021; 22:ijms22137028. [PMID: 34209958 PMCID: PMC8268687 DOI: 10.3390/ijms22137028] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 01/16/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease whose pathophysiology is largely unknown. Despite the fact that motor neuron (MN) death is recognized as the key event in ALS, astrocytes dysfunctionalities and neuroinflammation were demonstrated to accompany and probably even drive MN loss. Nevertheless, the mechanisms priming astrocyte failure and hyperactivation are still obscure. In this work, altered pathways and molecules in ALS astrocytes were unveiled by investigating the proteomic profile and the secreted metabolome of primary spinal cord astrocytes derived from transgenic ALS mouse model overexpressing the human (h)SOD1(G93A) protein in comparison with the transgenic counterpart expressing hSOD1(WT) protein. Here we show that ALS primary astrocytes are depleted of proteins-and of secreted metabolites-involved in glutathione metabolism and signaling. The observed increased activation of Nf-kB, Ebf1, and Plag1 transcription factors may account for the augmented expression of proteins involved in the proteolytic routes mediated by proteasome or endosome-lysosome systems. Moreover, hSOD1(G93A) primary astrocytes also display altered lipid metabolism. Our results provide novel insights into the altered molecular pathways that may underlie astrocyte dysfunctionalities and altered astrocyte-MN crosstalk in ALS, representing potential therapeutic targets to abrogate or slow down MN demise in disease pathogenesis.
Collapse
Affiliation(s)
- Roberto Stella
- Department of Chemistry, Istituto Zooprofilattico Sperimentale delle Venezie, 35020 Legnaro, Italy;
| | - Raphael Severino Bonadio
- Department of Biology and CRIBI Biotechnology Center, University of Padova, 35131 Padova, Italy; (R.S.B.); (S.C.)
| | - Stefano Cagnin
- Department of Biology and CRIBI Biotechnology Center, University of Padova, 35131 Padova, Italy; (R.S.B.); (S.C.)
- CIR-Myo Myology Center, University of Padova, 35131 Padova, Italy
| | | | - Alessandro Bertoli
- CNR—Neuroscience Institute, 35131 Padova, Italy;
- Padova Neuroscience Center, University of Padova, 35131 Padova, Italy
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
- Correspondence: (A.B.); (C.P.)
| | - Caterina Peggion
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
- Correspondence: (A.B.); (C.P.)
| |
Collapse
|