1
|
Ayyadurai VAS, Deonikar P, Kamm RD. A molecular systems architecture of neuromuscular junction in amyotrophic lateral sclerosis. NPJ Syst Biol Appl 2025; 11:27. [PMID: 40097438 PMCID: PMC11914587 DOI: 10.1038/s41540-025-00501-5] [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: 06/24/2024] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
A molecular systems architecture is presented for the neuromuscular junction (NMJ) in order to provide a framework for organizing complexity of biomolecular interactions in amyotrophic lateral sclerosis (ALS) using a systematic literature review process. ALS is a fatal motor neuron disease characterized by progressive degeneration of the upper and lower motor neurons that supply voluntary muscles. The neuromuscular junction contains cells such as upper and lower motor neurons, skeletal muscle cells, astrocytes, microglia, Schwann cells, and endothelial cells, which are implicated in pathogenesis of ALS. This molecular systems architecture provides a multi-layered understanding of the intra- and inter-cellular interactions in the ALS neuromuscular junction microenvironment, and may be utilized for target identification, discovery of single and combination therapeutics, and clinical strategies to treat ALS.
Collapse
Affiliation(s)
- V A Shiva Ayyadurai
- Systems Biology Group, CytoSolve Research Division, CytoSolve, Inc., Cambridge, MA, UK.
- Open Science Institute, International Center for Integrative Systems, Cambridge, MA, UK.
| | - Prabhakar Deonikar
- Systems Biology Group, CytoSolve Research Division, CytoSolve, Inc., Cambridge, MA, UK
- Open Science Institute, International Center for Integrative Systems, Cambridge, MA, UK
| | - Roger D Kamm
- Departments of Biological Engineering and Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, UK
| |
Collapse
|
2
|
Faller KME, Chaytow H, Gillingwater TH. Targeting common disease pathomechanisms to treat amyotrophic lateral sclerosis. Nat Rev Neurol 2025; 21:86-102. [PMID: 39743546 DOI: 10.1038/s41582-024-01049-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2024] [Indexed: 01/04/2025]
Abstract
The motor neuron disease amyotrophic lateral sclerosis (ALS) is a devastating condition with limited treatment options. The past few years have witnessed a ramping up of translational ALS research, offering the prospect of disease-modifying therapies. Although breakthroughs using gene-targeted approaches have shown potential to treat patients with specific disease-causing mutations, the applicability of such therapies remains restricted to a minority of individuals. Therapies targeting more general mechanisms that underlie motor neuron pathology in ALS are therefore of considerable interest. ALS pathology is associated with disruption to a complex array of key cellular pathways, including RNA processing, proteostasis, metabolism and inflammation. This Review details attempts to restore cellular homeostasis by targeting these pathways in order to develop effective, broadly-applicable ALS therapeutics.
Collapse
Affiliation(s)
- Kiterie M E Faller
- Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
- Royal (Dick) School of Veterinary Studies, University of Edinburgh, Midlothian, UK
| | - Helena Chaytow
- Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Edinburgh, UK
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK
| | - Thomas H Gillingwater
- Edinburgh Medical School, Biomedical Sciences, University of Edinburgh, Edinburgh, UK.
- Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
3
|
Weiner HL. Immune mechanisms and shared immune targets in neurodegenerative diseases. Nat Rev Neurol 2025; 21:67-85. [PMID: 39681722 DOI: 10.1038/s41582-024-01046-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/15/2024] [Indexed: 12/18/2024]
Abstract
The immune system plays a major part in neurodegenerative diseases. In some, such as multiple sclerosis, it is the primary driver of the disease. In others, such as Alzheimer disease, amyotrophic lateral sclerosis and Parkinson disease, it has an amplifying role. Immunotherapeutic approaches that target the adaptive and innate immune systems are being explored for the treatment of almost all neurological diseases, and the targets and approaches are often common across diseases. Microglia are the primary immune cells in the brain that contribute to disease pathogenesis, and are consequently a common immune target for therapy. Other therapeutic approaches target components of the peripheral immune system, such as regulatory T cells and monocytes, which in turn act within the CNS. This Review considers in detail how microglia, monocytes and T cells contribute to the pathogenesis of multiple sclerosis, Alzheimer disease, amyotrophic lateral sclerosis and Parkinson disease, and their potential as shared therapeutic targets across these diseases. The microbiome is also highlighted as an emerging therapeutic target that indirectly modulates the immune system. Therapeutic approaches being developed to target immune function in neurodegenerative diseases are discussed, highlighting how immune-based approaches developed to treat one disease could be applicable to multiple other neurological diseases.
Collapse
Affiliation(s)
- Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Yang J, Li W, Tian M, Zhang L, Du F, Li X, Liu Q, Li R, Li Z, Dong H, Liu Y. Cortical thickness correlated with peripheral inflammatory cytokines in amyotrophic lateral sclerosis. Front Neurosci 2025; 18:1514554. [PMID: 39840015 PMCID: PMC11747150 DOI: 10.3389/fnins.2024.1514554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/16/2024] [Indexed: 01/23/2025] Open
Abstract
Introduction Amyotrophic lateral sclerosis (ALS) is a rare, devastating neurodegenerative disease that affects upper and lower motor neurons, resulting in muscle atrophy, spasticity, hyperreflexia, and paralysis. Inflammation plays an important role in the development of ALS, and associated with rapid disease progression. Current observational studies indicate the thinning of cortical thickness in patients with ALS is associated with rapid disease progression and cognitive changes. However, the effects of inflammatory cytokines on cortical thickness in patients with ALS are unclear. Here, we investigated the relationship between inflammatory cytokines and cortical thickness in patients with ALS. Methods We evaluated 51 patients with ALS for inflammatory cytokines including interleukin (IL)-4, interferon (IFN)-α, IL-1β, IL-2, IL-5, IL-12, tumor necrosis factor (TNF)-α, IL-6, IL-10, IL-8, IL-17, and IFN-γ and analyzed the correlation between these indicators and the ALS functional rating scale-revised (ALSFRS-R) score or disease progression rate (ΔFS score). Twenty-six patients with ALS and 26 controls were studied using whole-cortex analysis, and post-hoc analyses were performed to examine the correlation between brain cortical thickness and ALSFRS-R or ΔFS scores. Results IL-4, IFN-α, IL-1β, and IL-2 levels were significantly correlated with ALSFRS-R scores, and the IL-2 level was significantly correlated with ΔFS scores. After controlling for age and sex, the ALS group had thinner cortexes in multiple clusters across the brain than the control group. Further analyses revealed that cortical thickness in the right superior temporal and lingual gyrus regions was inversely correlated with ΔFS scores. There was a significant positive correlation between the clusters in the right lingual cortex and IL-2 level. Conclusion These results suggest cortical thickness was reduced in patients with ALS in motor and non-motor cortical areas. Inflammatory factors (especially IL-2) were correlated with cortical thickness, and both were related to the disease progression rate, suggesting IL-2 plays an important role in ALS.
Collapse
Affiliation(s)
- Jing Yang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Wenyi Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Mei Tian
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Lei Zhang
- Department of Emergency, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Fengping Du
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Xin Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Qi Liu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Rui Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Zhenzhong Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Hui Dong
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Yaling Liu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| |
Collapse
|
5
|
Zeng Y, Guo R, Cao S, Chavarria Gonzalez S, Pang K, Liu C, Yang H. Mendelian randomization study supports relative carbohydrate intake as an independent risk factor for amyotrophic lateral sclerosis. Nutr Neurosci 2025; 28:116-124. [PMID: 38781481 DOI: 10.1080/1028415x.2024.2352196] [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] [Indexed: 05/25/2024]
Abstract
OBJECTIVES Observational studies suggested a potential correlation between dietary intake and amyotrophic lateral sclerosis (ALS), but conflicting findings exist and causality remains unclear. Here, we performed a Mendelian randomization (MR) analysis to evaluate the causal impact of relative intake of (i) carbohydrate, (ii) fat, and (iii) protein on ALS risk. METHODS The genome-wide association summary statistics of three dietary macronutrient intake traits and ALS were obtained. Initially, forward and reverse univariable MR (UVMR) analysis were conducted using the inverse variance weighted (IVW) method as the primary approach, supplemented by MR-Egger, weighted median, and maximum likelihood. Subsequently, multivariable MR (MVMR) analysis was performed to assess the independent causal effects of each dietary. Additionally, diverse sensitivity tests were conducted to evaluate the reliability of the MR analyses. RESULTS The forward UVMR analysis conducted by IVW indicated that relative carbohydrate intake significantly increased ALS risk. Furthermore, results from three other MR methods paralleled those from IVW. However, the other two dietary intake traits did not have a causative impact on ALS risk. The reverse UVMR analysis indicated that ALS did not causatively influence the three dietary intake traits. The MVMR analysis showed that after adjusting for the effects of the other two dietary intake traits, relative carbohydrate intake independently and significantly increased ALS risk. Sensitivity tests indicated no significant heterogeneity or horizontal pleiotropy. DISCUSSION MR analysis supported relative carbohydrate independently increasing ALS risk. Nevertheless, further validation of this finding in future large cohorts is required.
Collapse
Affiliation(s)
- Youjie Zeng
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Ren Guo
- Department of Pharmacy, Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Si Cao
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-XIANGYA, Changsha, People's Republic of China
| | - Sarel Chavarria Gonzalez
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Ke Pang
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Chunxia Liu
- Department of Anesthesiology, Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Heng Yang
- Department of Neurology, Third Xiangya Hospital, Central South University, Changsha, People's Republic of China
| |
Collapse
|
6
|
Hossainpour H, Khazaei S, Mahmoudi H. The Effect of Probiotics and Microbiota on Nervous System and Mental Illnesses. Cent Nerv Syst Agents Med Chem 2025; 25:109-122. [PMID: 39313893 DOI: 10.2174/0118715249315760240905043415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 07/08/2024] [Accepted: 07/29/2024] [Indexed: 09/25/2024]
Abstract
The microbiota that inhabits the gastrointestinal tract has been linked to various gastrointestinal and non-gastrointestinal disorders. Scientists have been studying how the bacteria in our intestines have an effect on our brain and nervous system. This connection is called the "microbiota-gut-brain axis". Given the capacity of probiotics, which are live non-pathogenic microorganisms, to reinstate the normal microbial population within the host and confer advantages, their potential impact has been subjected to scrutiny with regard to neurological and mental conditions. Material sourced for this review included peer-reviewed literature annotated in the PubMed, Web of Science, Scopus, and Google Scholar databases. The result has indicated the integration of probiotics into a child's diet to enhance the neuro-behavioral symptoms. Notwithstanding this, the current data set has been found to be insufficient and inconclusive. The potential utility of probiotics for the prevention or treatment of neurologic and mental disorders has become a subject of substantial interest.
Collapse
Affiliation(s)
- Hadi Hossainpour
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Microbiology, School of Medicine, Student Research Committee, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Salman Khazaei
- Department of Epidemiology, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hassan Mahmoudi
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Microbiology, Nahavand School of Allied Medical Sciences, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
7
|
Fang K. Modulation of the central nervous system immune response and neuroinflammation via Wnt signaling in health and neurodegenerative diseases. IBRAIN 2024; 10:462-476. [PMID: 39691422 PMCID: PMC11649390 DOI: 10.1002/ibra.12185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 11/02/2024] [Accepted: 11/07/2024] [Indexed: 12/19/2024]
Abstract
The immune response in the central nervous system (CNS) is a highly specialized and tightly regulated process essential for maintaining neural health and protecting against pathogens and injuries. The primary immune cells within the CNS include microglia, astrocytes, T cells, and B cells. They work together, continuously monitor the CNS environment for signs of infection, injury, or disease, and respond by phagocytosing debris, releasing cytokines, and recruiting other immune cells. In addition to providing neuroprotection, these immune responses must be carefully balanced to prevent excessive inflammation that can lead to neuronal damage and contribute to neurodegenerative diseases. Dysregulated immune responses in the CNS are implicated in various neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and amyotrophic lateral sclerosis. Wnt signaling is a crucial pathway in the CNS that regulates various cellular processes critical for brain development, function, and maintenance. Despite enhancing immune responses in the health CNS, dysregulated Wnt signaling exacerbates neuroinflammation in the neurodegenerative brains. This review summarized the role of Wnt signaling in regulating immune response under different conditions. We then examined the role of immune response in healthy brains and during the development of neurodegenerative diseases. We also discussed therapeutic intervention in various neurodegenerative diseases through the modulation of the Wnt signaling pathway and neuroinflammation and highlighted challenges and limitations in current clinical trials.
Collapse
Affiliation(s)
- Kevin Fang
- Living Systems InstituteUniversity ExeterExeterUK
| |
Collapse
|
8
|
Nelson RB, Rose KN, Menniti FS, Zorn SH. Hiding in plain sight: Do recruited dendritic cells surround amyloid plaques in Alzheimer's disease? Biochem Pharmacol 2024; 228:116258. [PMID: 38705533 DOI: 10.1016/j.bcp.2024.116258] [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: 02/23/2024] [Revised: 04/18/2024] [Accepted: 05/02/2024] [Indexed: 05/07/2024]
Abstract
Over the past decade, human genome-wide association and expression studies have strongly implicated dysregulation of the innate immune system in the pathogenesis of Alzheimer's disease (AD). Single cell mRNA sequencing studies have identified innate immune cell subtypes that are minimally present in normal healthy brain, but whose numbers greatly increase in association with AD pathology. These AD pathology-associated immune cells are putatively the locus for the immune-related AD risk. While the prevailing view is that these immune cells arise from transformation of resident brain microglia, studies across several decades and using multiple techniques and strategies suggest instead that the pathology-associated immune cells are bone-marrow derived hematopoietic cells that are recruited into brain. We critically review this translational literature, emphasizing the strengths and limitations of techniques used to address recruitment and the experimental designs employed. We conclude that the aggregate evidence points toward recruitment into brain of innate immune cells of the myeloid dendritic cell lineage. Recruitment of dendritic cells and their role in AD pathogenesis has broad implications for our understanding of the etiology and pathobiology of AD that impact the strategies to develop new, immune system-targeted therapeutics for this devastating disease.
Collapse
Affiliation(s)
- Robert B Nelson
- MindImmune Therapeutics, Inc., Kingston, RI; George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI; Dept of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI.
| | - Kenneth N Rose
- MindImmune Therapeutics, Inc., Kingston, RI; Dept of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI
| | - Frank S Menniti
- MindImmune Therapeutics, Inc., Kingston, RI; George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI; Dept of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI
| | - Stevin H Zorn
- MindImmune Therapeutics, Inc., Kingston, RI; George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI; Dept of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, RI
| |
Collapse
|
9
|
Dandamudi BJ, Dimaano KAM, Shah N, AlQassab O, Al-Sulaitti Z, Nelakuditi B, Mohammed L. Neurodegenerative Disorders and the Gut-Microbiome-Brain Axis: A Literature Review. Cureus 2024; 16:e72427. [PMID: 39588438 PMCID: PMC11588320 DOI: 10.7759/cureus.72427] [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: 09/03/2024] [Accepted: 10/26/2024] [Indexed: 11/27/2024] Open
Abstract
Neurodegenerative diseases are severe, age-related conditions with complex etiologies that result in significant morbidity and mortality. The gut microbiome, a dynamic symbiotic environment comprising commensal organisms, represents the largest reservoir of these organisms within the human body. It produces short-chain fatty acids, endogenous signals, and neuroactive compounds, which can modulate neuronal function, plasticity, and behavior. Emerging evidence suggests that the gut microbiome plays a pivotal role in neurodevelopment, aging, and brain diseases, including Alzheimer's disease, Parkinson's disease, and stroke. Communication between the gut and brain occurs through a bidirectional channel known as the gut-microbiome-brain axis, which is being explored for therapeutic potential in neurodegenerative disorders. This literature review was conducted through a comprehensive search of five electronic databases - PubMed, Scopus, Ovid Medline, Cochrane Review, and Google Scholar - from inception to June 2024, focusing on English-language studies. Keywords included "gut-brain axis", "microbiome dysbiosis", "neurodegeneration", and disorder-specific terms such as "Alzheimer's disease" and "Parkinson's disease", paired with "gut microbiome". The review examines current knowledge on the relationship between gut microbiota and neurodegenerative disorders, emphasizing potential mechanisms and therapeutic options. Results indicate that gut dysbiosis, characterized by microbial imbalance, is intricately associated with neurodegenerative disease pathogenesis by influencing immune responses, increasing blood-brain barrier permeability, and generating neurotoxic metabolites. Therapeutic approaches targeting the gut microbiome, including probiotics, prebiotics, and fecal microbiota transplantation, show promise in restoring microbial balance and slowing disease progression. However, further research is essential to validate these findings and develop effective clinical interventions.
Collapse
Affiliation(s)
- Bindu Jyothi Dandamudi
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Kathrina Antheia M Dimaano
- Obstetrics and Gynecology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Nensi Shah
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Osamah AlQassab
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Zainab Al-Sulaitti
- Obstetrics and Gynecology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Bhavana Nelakuditi
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Lubna Mohammed
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
10
|
Yang J, Tian M, Zhang L, Xin C, Huo J, Liu Q, Dong H, Li R, Liu Y. Assessment of Rab geranylgeranyltransferase subunit beta in amyotrophic lateral sclerosis. Front Neurol 2024; 15:1447461. [PMID: 39224887 PMCID: PMC11366579 DOI: 10.3389/fneur.2024.1447461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Geranylgeranyltransferase Subunit Beta (RABGGTB) was expressed at higher levels in patients with Amyotrophic lateral sclerosis (ALS) compared with healthy controls. This study aims to observe the expression of RABGGTB in different cells from patients with ALS and different diseases. Methods In this case-control study, we collected peripheral blood from patients with ALS and healthy controls, and compared the expression of RABGGTB in natural killer cells (NK), T cells and B cells between patients with ALS and healthy controls by flow cytometry. And compared the expression of RABGGTB in monocytes and monocyte-derived macrophages from patients with ALS, Parkinson's disease (PD), acute cerebrovascular disease (ACVD), and healthy controls by flow cytometry and immunofluorescence. Then flow cytometry was used to detect the expression of RABGGTB in monocytes from SOD1G93A mice and WT mice. Results The expression of RABGGTB was not significantly changed in NK cells, cytotoxic T cells (CTL), helper T cells (Th), regulatory T cells (Treg), and B cells from patients with ALS compared to healthy controls. And the expression of RABGGTB in monocytes and monocyte-derived macrophages was higher in the ALS group than in the PD, ACVD and control group. The expression of RABGGTB was significantly higher in monocytes of SOD1G93A mice compared to WT mice. Conclusion These findings suggest that RABGGTB expression was increased in monocytes and monocyte-derived macrophages from patients with ALS, not in NK, CTL, Th, Treg, and B cells. Future studies are needed to find the clinical implication of RABGGTB in ALS.
Collapse
Affiliation(s)
- Jing Yang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Mei Tian
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Lei Zhang
- Department of Emergency, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Cheng Xin
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Jia Huo
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Qi Liu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Hui Dong
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Rui Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| | - Yaling Liu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
- The Key Laboratory of Neurology, Hebei Medical University, Ministry of Education, Shijiazhuang, Hebei, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, Hebei, China
| |
Collapse
|
11
|
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
|
12
|
Yang T, Wei Q, Li C, Ou R, Lin J, Cheng Y, Xiao Y, Shang H. Peripheral immunity involvement in the cognitive impairment of sporadic amyotrophic lateral sclerosis. Front Neurol 2024; 15:1405275. [PMID: 38882692 PMCID: PMC11176427 DOI: 10.3389/fneur.2024.1405275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/15/2024] [Indexed: 06/18/2024] Open
Abstract
Background Recent research has indicated the significance of immune activation in amyotrophic lateral sclerosis (ALS). However, the impact of peripheral immunity on cognitive impairment in sporadic ALS remains poorly characterized. Therefore, we aim to assess the relationship between peripheral immune parameters and cognitive impairment in patients with sporadic ALS. Methods A case-control study involving 289 patients with sporadic ALS was conducted. All participants underwent cognitive assessment and measurements of blood immune parameters. The main outcomes included adjusted odds ratios (ORs) in multivariate logistic regression analysis and adjusted coefficients in a multivariate linear regression model. Sensitivity analysis was performed with stratification by the King's clinical stage. Results Cognitive impairment was observed in 98 (33.9%) patients. Higher counts of leukocyte (OR, 0.53; 95% CI, 0.29 to 0.95; p = 0.03), neutrophil (OR, 0.48; 95% CI, 0.26 to 0.88; p = 0.02), and monocyte (OR, 0.33; 95% CI, 0.18 to 0.60; p < 0.001) were significantly associated with better cognitive preformence in sporadic ALS, particularly among patients in King's clinical stages 1 and 2. Conversely, a higher percentage of CD4+ T cells was linked to an increased risk of cognitive impairment (OR, 2.79; 95% CI, 1.52 to 5.09; p = 0.001), particularly evident in patients in King's clinical stage 3. Conclusion These results highlight the involvement of peripheral immunity in the cognitive impairment of sporadic ALS and suggest dynamic and intricate roles that vary across disease stages. Elucidating the links between immunity and ALS sheds light on the pathophysiological mechanisms underlying this fatal neurodegenerative disorder and informs potential immunotherapeutic strategies.
Collapse
Affiliation(s)
- Tianmi Yang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qianqian Wei
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chunyu Li
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruwei Ou
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junyu Lin
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yangfan Cheng
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Xiao
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huifang Shang
- Department of Neurology, Laboratory of Neurodegenerative Disorders, Rare Diseases Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
13
|
Pandya VA, Patani R. The role of glial cells in amyotrophic lateral sclerosis. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2024; 176:381-450. [PMID: 38802179 DOI: 10.1016/bs.irn.2024.04.005] [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: 05/29/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) has traditionally been considered a neuron-centric disease. This view is now outdated, with increasing recognition of cell autonomous and non-cell autonomous contributions of central and peripheral nervous system glia to ALS pathomechanisms. With glial research rapidly accelerating, we comprehensively interrogate the roles of astrocytes, microglia, oligodendrocytes, ependymal cells, Schwann cells and satellite glia in nervous system physiology and ALS-associated pathology. Moreover, we highlight the inter-glial, glial-neuronal and inter-system polylogue which constitutes the healthy nervous system and destabilises in disease. We also propose classification based on function for complex glial reactive phenotypes and discuss the pre-requisite for integrative modelling to advance translation. Given the paucity of life-enhancing therapies currently available for ALS patients, we discuss the promising potential of harnessing glia in driving ALS therapeutic discovery.
Collapse
Affiliation(s)
- Virenkumar A Pandya
- University College London Medical School, London, United Kingdom; The Francis Crick Institute, London, United Kingdom.
| | - Rickie Patani
- The Francis Crick Institute, London, United Kingdom; Department of Neuromuscular Diseases, University College London Queen Square Institute of Neurology, Queen Square, London, United Kingdom.
| |
Collapse
|
14
|
Murdock BJ, Zhao B, Pawlowski KD, Famie JP, Piecuch CE, Webber-Davis IF, Teener SJ, Feldman EL, Zhao L, Goutman SA. Peripheral Immune Profiles Predict ALS Progression in an Age- and Sex-Dependent Manner. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200241. [PMID: 38626361 PMCID: PMC11087030 DOI: 10.1212/nxi.0000000000200241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 02/12/2024] [Indexed: 04/18/2024]
Abstract
BACKGROUND AND OBJECTIVES Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease whose pathobiology associates with peripheral blood immune cell levels and activation patterns in an age and sex-dependent manner. This study's objective was to identify immune profile associations with ALS progression, whether the associations are age and sex-specific, and whether immune profiles can predict a future disease course. METHODS Flow cytometry immune profiles (a combination of 22 peripheral blood immune markers) were generated for 241 participants with ALS and linked to ALS progression, using progression-free survival, which is a composite combining the revised ALS Functional Rating Scale and survival. Participants were first grouped by immune profiles using unsupervised hierarchical clustering, and clusters were associated with subsequent progression-free survival. Next, individual immune markers were associated with progression-free survival using least absolute shrinkage and selection operator-Cox regression. Analyses were stratified by age and sex to identify demographic-specific immune mechanisms. Finally, random forest determined the predictive power of immune profiles on ALS progression in the whole population and again stratified by age and sex. RESULTS Progression-free survival differed between clusters of participants with similar immune profiles, particularly reduced natural killer (NK)-cell activation associated with slower progression. Individual markers such as neutrophil levels and NK-cell NKp46 expression associated with faster ALS progression while overall NK-cell levels and NK-cell subpopulations associated with slower progression; the strength of these associations varied by age and sex. Adding these immune markers to prediction models dramatically increased short-term prediction compared with routine clinical prognostic variables alone, and the addition of NK-cell markers further improved the prediction accuracy in female participants. DISCUSSION Specific immune profiles likely contribute to ALS progression in an age and sex-dependent manner, and peripheral immune markers enhance the prediction of short-term clinical outcomes. These findings suggest a complex milieu of immune profiles associated with ALS progression, and more detailed immunophenotyping in ALS will facilitate personalized immunotherapeutics in ALS.
Collapse
Affiliation(s)
- Benjamin J Murdock
- From the Department of Neurology (B.J.M., K.D.P., J.P.F., C.E.P., I.F.W.-D., S.J.T., E.L.F., S.A.G.); and School of Public Health (B.Z., L.Z.), Biostatistics Department, University of Michigan, Ann Arbor
| | - Bangyao Zhao
- From the Department of Neurology (B.J.M., K.D.P., J.P.F., C.E.P., I.F.W.-D., S.J.T., E.L.F., S.A.G.); and School of Public Health (B.Z., L.Z.), Biostatistics Department, University of Michigan, Ann Arbor
| | - Kristen D Pawlowski
- From the Department of Neurology (B.J.M., K.D.P., J.P.F., C.E.P., I.F.W.-D., S.J.T., E.L.F., S.A.G.); and School of Public Health (B.Z., L.Z.), Biostatistics Department, University of Michigan, Ann Arbor
| | - Joshua P Famie
- From the Department of Neurology (B.J.M., K.D.P., J.P.F., C.E.P., I.F.W.-D., S.J.T., E.L.F., S.A.G.); and School of Public Health (B.Z., L.Z.), Biostatistics Department, University of Michigan, Ann Arbor
| | - Caroline E Piecuch
- From the Department of Neurology (B.J.M., K.D.P., J.P.F., C.E.P., I.F.W.-D., S.J.T., E.L.F., S.A.G.); and School of Public Health (B.Z., L.Z.), Biostatistics Department, University of Michigan, Ann Arbor
| | - Ian F Webber-Davis
- From the Department of Neurology (B.J.M., K.D.P., J.P.F., C.E.P., I.F.W.-D., S.J.T., E.L.F., S.A.G.); and School of Public Health (B.Z., L.Z.), Biostatistics Department, University of Michigan, Ann Arbor
| | - Samuel J Teener
- From the Department of Neurology (B.J.M., K.D.P., J.P.F., C.E.P., I.F.W.-D., S.J.T., E.L.F., S.A.G.); and School of Public Health (B.Z., L.Z.), Biostatistics Department, University of Michigan, Ann Arbor
| | - Eva L Feldman
- From the Department of Neurology (B.J.M., K.D.P., J.P.F., C.E.P., I.F.W.-D., S.J.T., E.L.F., S.A.G.); and School of Public Health (B.Z., L.Z.), Biostatistics Department, University of Michigan, Ann Arbor
| | - Lili Zhao
- From the Department of Neurology (B.J.M., K.D.P., J.P.F., C.E.P., I.F.W.-D., S.J.T., E.L.F., S.A.G.); and School of Public Health (B.Z., L.Z.), Biostatistics Department, University of Michigan, Ann Arbor
| | - Stephen A Goutman
- From the Department of Neurology (B.J.M., K.D.P., J.P.F., C.E.P., I.F.W.-D., S.J.T., E.L.F., S.A.G.); and School of Public Health (B.Z., L.Z.), Biostatistics Department, University of Michigan, Ann Arbor
| |
Collapse
|
15
|
Arora H, Javed B, Kutikuppala LVS, Chaurasia M, Khullar K, Kannan S, Golla V. ST2 levels and neurodegenerative diseases: is this a significant relation? Ann Med Surg (Lond) 2024; 86:2812-2817. [PMID: 38694387 PMCID: PMC11060292 DOI: 10.1097/ms9.0000000000001939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 02/29/2024] [Indexed: 05/04/2024] Open
Abstract
Interleukin-33 (IL-33), belonging to the interleukin-1 cytokine family, has a decoy receptor soluble ST2 (sST2). IL-33 is found in oligodendrocytes and astrocytes and is involved in central nervous system healing and repair, whereas ST2 is found in microglia and astrocytes. Some studies have found a link between changes in the IL-33/ST2 pathway and neurodegenerative disorders. This review article investigates the relationship between the interleukin-33 (IL-33)/ST2 pathway and neurodegenerative disorders. It was discovered that soluble st2 levels were increased. Furthermore, IL-33 levels were found to be lower in many neurodegenerative diseases such as Alzheimer's and amyotrophic lateral sclerosis (ALS). The association with other disorders, such as ankylosing spondylitis, multiple sclerosis, and systemic lupus erythematosus (SLE), was also observed. Various studies suggest that ST2/IL-33 signalling may be pivotal in the disease modulation of neurodegenerative disorders. The serum sST2 level test can be useful in determining the inflammatory status and severity of illness in many neurodegenerative disorders. In this review, we will discuss recent findings concerning the interleukin-33 (IL-33)/ST2 pathway and its role in the diagnosis and treatment of diseases with neurodegeneration.
Collapse
Affiliation(s)
- Himanshu Arora
- Department of General Medicine, Netaji Subhash Chandra Bose Subharti Medical College, Meerut, Uttar Pradesh
| | - Binish Javed
- Atal Bihari Vajpayee Institute of Medical Sciences & Dr. Ram Manohar Lohia Hospital, New Delhi
| | | | - Mayuri Chaurasia
- National Institute of Medical Sciences and Research, Jaipur, Rajasthan
| | | | - Shreevikaa Kannan
- Department of General Medicine Tbilisi State Medical University, Tbilisi, Georgia
| | - Varshitha Golla
- Department of General Medicine, International School of Medicine (ISM), Bishkek, Kyrgyzstan
| |
Collapse
|
16
|
Caggiano C, Morselli M, Qian X, Celona B, Thompson M, Wani S, Tosevska A, Taraszka K, Heuer G, Ngo S, Steyn F, Nestor P, Wallace L, McCombe P, Heggie S, Thorpe K, McElligott C, English G, Henders A, Henderson R, Lomen-Hoerth C, Wray N, McRae A, Pellegrini M, Garton F, Zaitlen N. Tissue informative cell-free DNA methylation sites in amyotrophic lateral sclerosis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.04.08.24305503. [PMID: 38645132 PMCID: PMC11030489 DOI: 10.1101/2024.04.08.24305503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
Cell-free DNA (cfDNA) is increasingly recognized as a promising biomarker candidate for disease monitoring. However, its utility in neurodegenerative diseases, like amyotrophic lateral sclerosis (ALS), remains underexplored. Existing biomarker discovery approaches are tailored to a specific disease context or are too expensive to be clinically practical. Here, we address these challenges through a new approach combining advances in molecular and computational technologies. First, we develop statistical tools to select tissue-informative DNA methylation sites relevant to a disease process of interest. We then employ a capture protocol to select these sites and perform targeted methylation sequencing. Multi-modal information about the DNA methylation patterns are then utilized in machine learning algorithms trained to predict disease status and disease progression. We applied our method to two independent cohorts of ALS patients and controls (n=192). Overall, we found that the targeted sites accurately predicted ALS status and replicated between cohorts. Additionally, we identified epigenetic features associated with ALS phenotypes, including disease severity. These findings highlight the potential of cfDNA as a non-invasive biomarker for ALS.
Collapse
Affiliation(s)
- C Caggiano
- Department of Neurology, UCLA, Los Angeles, California
- Institute of Genomic Health, Icahn School of Medicine at Mt Sinai, New York, New York
| | - M Morselli
- Department of Molecular, Cell, and Developmental Biology, UCLA; Los Angeles, California
- Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - X Qian
- Institute for Molecular Biology, University of Queensland, Brisbane, Australia
| | - B Celona
- Cardiovascular Research Institute, UCSF, San Francisco, California
| | - M Thompson
- Department of Neurology, UCLA, Los Angeles, California
- Systems and Synthetic Biology, Centre for Genomic Regulation, Barcelona, Spain
| | - S Wani
- Cardiovascular Research Institute, UCSF, San Francisco, California
| | - A Tosevska
- Department of Molecular, Cell, and Developmental Biology, UCLA; Los Angeles, California
- Department of Internal Medicine III, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
| | - K Taraszka
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - G Heuer
- Computational and Systems Biology Interdepartmental Program, UCLA, Los Angeles, California
| | - S Ngo
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Australia
- Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - F Steyn
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - P Nestor
- Queensland Brain Institute, Unviversity of Queensland, Brisbane, Australia
- Mater Public Hospital, Brisbane, Australia
| | - L Wallace
- Institute for Molecular Biology, University of Queensland, Brisbane, Australia
| | - P McCombe
- Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - S Heggie
- Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - K Thorpe
- Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | | | - G English
- Institute for Molecular Biology, University of Queensland, Brisbane, Australia
| | - A Henders
- Institute for Molecular Biology, University of Queensland, Brisbane, Australia
| | - R Henderson
- Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - C Lomen-Hoerth
- Department of Neurology, UCSF, San Francisco, California
| | - N Wray
- Institute for Molecular Biology, University of Queensland, Brisbane, Australia
| | - A McRae
- Institute for Molecular Biology, University of Queensland, Brisbane, Australia
| | - M Pellegrini
- Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - F Garton
- Institute for Molecular Biology, University of Queensland, Brisbane, Australia
| | - N Zaitlen
- Department of Neurology, UCLA, Los Angeles, California
- Department of Human Genetics, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
17
|
Moțățăianu A, Andone S, Stoian A, Bălașa R, Huțanu A, Sărmășan E. A Potential Role of Interleukin-5 in the Pathogenesis and Progression of Amyotrophic Lateral Sclerosis: A New Molecular Perspective. Int J Mol Sci 2024; 25:3782. [PMID: 38612591 PMCID: PMC11011909 DOI: 10.3390/ijms25073782] [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: 02/26/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Cumulative data suggest that neuroinflammation plays a prominent role in amyotrophic lateral sclerosis (ALS) pathogenesis. The purpose of this work was to assess if patients with ALS present a specific peripheral cytokine profile and if it correlates with neurological disability assessed by ALSFRS-R, the rate of disease progression, and the pattern of disease progression (horizontal spreading [HSP] versus vertical spreading [VSP]). We determined the levels of 15 cytokines in the blood of 59 patients with ALS and 40 controls. We identified a positive correlation between levels of pro-inflammatory cytokines (interleukin [IL]-17F, IL-33, IL-31) and the age of ALS patients, as well as a positive correlation between IL-12p/70 and survival from ALS onset and ALS diagnosis. Additionally, there was a positive correlation between the ALSFRS-R score in the upper limb and respiratory domain and IL-5 levels. In our ALS cohort, the spreading pattern was 42% horizontal and 58% vertical, with patients with VSP showing a faster rate of ALS progression. Furthermore, we identified a negative correlation between IL-5 levels and the rate of disease progression, as well as a positive correlation between IL-5 and HSP of ALS. To the best of our knowledge, this is the first study reporting a "protective" role of IL-5 in ALS.
Collapse
Affiliation(s)
- Anca Moțățăianu
- 1st Neurology Clinic, Mures County Clinical Emergency Hospital, 540136 Targu Mures, Romania; (A.M.); (S.A.); (A.S.); (R.B.); (E.S.)
- Department of Neurology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Sebastian Andone
- 1st Neurology Clinic, Mures County Clinical Emergency Hospital, 540136 Targu Mures, Romania; (A.M.); (S.A.); (A.S.); (R.B.); (E.S.)
- Department of Neurology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Adina Stoian
- 1st Neurology Clinic, Mures County Clinical Emergency Hospital, 540136 Targu Mures, Romania; (A.M.); (S.A.); (A.S.); (R.B.); (E.S.)
- Department of Pathophysiology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Rodica Bălașa
- 1st Neurology Clinic, Mures County Clinical Emergency Hospital, 540136 Targu Mures, Romania; (A.M.); (S.A.); (A.S.); (R.B.); (E.S.)
- Department of Neurology, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Adina Huțanu
- Department of Laboratory Medicine, Mures County Clinical Emergency Hospital, 540136 Targu Mures, Romania
- Department of Laboratory Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania
| | - Emanuela Sărmășan
- 1st Neurology Clinic, Mures County Clinical Emergency Hospital, 540136 Targu Mures, Romania; (A.M.); (S.A.); (A.S.); (R.B.); (E.S.)
| |
Collapse
|
18
|
Komine O, Ohnuma S, Hinohara K, Hara Y, Shimada M, Akashi T, Watanabe S, Sobue A, Kawade N, Ogi T, Yamanaka K. Genetic background variation impacts microglial heterogeneity and disease progression in amyotrophic lateral sclerosis model mice. iScience 2024; 27:108872. [PMID: 38318390 PMCID: PMC10839647 DOI: 10.1016/j.isci.2024.108872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 11/07/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Recent single-cell analyses have revealed the complexity of microglial heterogeneity in brain development, aging, and neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS). Disease-associated microglia (DAMs) have been identified in ALS mice model, but their role in ALS pathology remains unclear. The effect of genetic background variations on microglial heterogeneity and functions remains unknown. Herein, we established and analyzed two mice models of ALS with distinct genetic backgrounds of C57BL/6 and BALB/c. We observed that the change in genetic background from C57BL/6 to BALB/c affected microglial heterogeneity and ALS pathology and its progression, likely due to the defective induction of neurotrophic factor-secreting DAMs and impaired microglial survival. Single-cell analyses of ALS mice revealed new markers for each microglial subtype and a possible association between microglial heterogeneity and systemic immune environments. Thus, we highlighted the role of microglia in ALS pathology and importance of genetic background variations in modulating microglial functions.
Collapse
Affiliation(s)
- Okiru Komine
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Nagoya University, Nagoya, Aichi, Japan
| | - Syuhei Ohnuma
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Nagoya University, Nagoya, Aichi, Japan
| | - Kunihiko Hinohara
- Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Institute for Advanced Research, Nagoya University, Nagoya, Aichi, Japan
- Center for 5D Cell Dynamics, Nagoya University, Nagoya, Aichi, Japan
| | - Yuichiro Hara
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
- Department of Human Genetics and Molecular Biology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Mayuko Shimada
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
- Department of Human Genetics and Molecular Biology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Tomohiro Akashi
- Center for 5D Cell Dynamics, Nagoya University, Nagoya, Aichi, Japan
- Center for Neurological Disease and Cancer, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Seiji Watanabe
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Nagoya University, Nagoya, Aichi, Japan
| | - Akira Sobue
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Nagoya University, Nagoya, Aichi, Japan
- Medical Interactive Research and Academia Industry Collaboration Center, Research Institute of Environmental Medicine, Nagoya University, Aichi, Japan
| | - Noe Kawade
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Nagoya University, Nagoya, Aichi, Japan
| | - Tomoo Ogi
- Department of Genetics, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
- Department of Human Genetics and Molecular Biology, Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
- Institute for Glyco-core Research (iGCORE), Nagoya University, Aichi, Japan
- Center for One Medicine Innovative Translational Research (COMIT), Nagoya University, Aichi, Japan
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Aichi, Japan
- Department of Neuroscience and Pathobiology, Nagoya University Graduate School of Medicine, Nagoya University, Nagoya, Aichi, Japan
- Institute for Glyco-core Research (iGCORE), Nagoya University, Aichi, Japan
- Center for One Medicine Innovative Translational Research (COMIT), Nagoya University, Aichi, Japan
| |
Collapse
|
19
|
Sutter PA, Lavoie ER, Lombardo ET, Pinter MK, Crocker SJ. Emerging Role of Astrocyte-Derived Extracellular Vesicles as Active Participants in CNS Neuroimmune Responses. Immunol Invest 2024; 53:26-39. [PMID: 37981468 PMCID: PMC11472422 DOI: 10.1080/08820139.2023.2281621] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2023]
Abstract
Astrocyte-derived extracellular vesicles (ADEVs) have garnered attention as a fundamental mechanism of intercellular communication in health and disease. In the context of neurological diseases, for which prodromal diagnosis would be advantageous, ADEVs are also being explored for their potential utility as biomarkers. In this review, we provide the current state of data supporting our understanding on the manifold roles of ADEVs in several common neurological disorders. We also discuss these findings from a unique emerging perspective that ADEVs represent a means by which the central nervous system may broadcast influence over other systems in the body to affect neuroinflammatory processes, with both dual potential to either propagate illness or restore health and homeostasis.
Collapse
Affiliation(s)
- Pearl A. Sutter
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Erica R. Lavoie
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Evan T. Lombardo
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Meghan K. Pinter
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030
| | - Stephen J. Crocker
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT 06030
| |
Collapse
|
20
|
Dey B, Kumar A, Patel AB. Pathomechanistic Networks of Motor System Injury in Amyotrophic Lateral Sclerosis. Curr Neuropharmacol 2024; 22:1778-1806. [PMID: 37622689 PMCID: PMC11284732 DOI: 10.2174/1570159x21666230824091601] [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: 04/21/2023] [Revised: 05/25/2023] [Accepted: 06/06/2023] [Indexed: 08/26/2023] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is the most common, adult-onset, progressive motor neurodegenerative disorder that results in death within 3 years of the clinical diagnosis. Due to the clinicopathological heterogeneity, any reliable biomarkers for diagnosis or prognosis of ALS have not been identified till date. Moreover, the only three clinically approved treatments are not uniformly effective in slowing the disease progression. Over the last 15 years, there has been a rapid advancement in research on the complex pathomechanistic landscape of ALS that has opened up new avenues for successful clinical translation of targeted therapeutics. Multiple studies suggest that the age-dependent interaction of risk-associated genes with environmental factors and endogenous modifiers is critical to the multi-step process of ALS pathogenesis. In this review, we provide an updated discussion on the dysregulated cross-talk between intracellular homeostasis processes, the unique molecular networks across selectively vulnerable cell types, and the multisystemic nature of ALS pathomechanisms. Importantly, this work highlights the alteration in epigenetic and epitranscriptomic landscape due to gene-environment interactions, which have been largely overlooked in the context of ALS pathology. Finally, we suggest that precision medicine research in ALS will be largely benefitted from the stratification of patient groups based on the clinical phenotype, onset and progression, genome, exposome, and metabolic identities.
Collapse
Affiliation(s)
- Bedaballi Dey
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad 500007, Telangana, India
- AcSIR-Academy of Scientific and Innovative Research, Ghaziabad 201002, Uttar Pradesh, India
| | - Arvind Kumar
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad 500007, Telangana, India
- AcSIR-Academy of Scientific and Innovative Research, Ghaziabad 201002, Uttar Pradesh, India
| | - Anant Bahadur Patel
- CSIR-Centre for Cellular and Molecular Biology (CSIR-CCMB), Hyderabad 500007, Telangana, India
- AcSIR-Academy of Scientific and Innovative Research, Ghaziabad 201002, Uttar Pradesh, India
| |
Collapse
|
21
|
Yang J, Liu T, Zhang L, Li X, Du FP, Liu Q, Dong H, Liu Y. Eosinophils at diagnosis are elevated in amyotrophic lateral sclerosis. Front Neurol 2023; 14:1289467. [PMID: 38187158 PMCID: PMC10768070 DOI: 10.3389/fneur.2023.1289467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
Introduction Amyotrophic lateral sclerosis (ALS) is a rare, devastating neurodegenerative disease that affects upper and lower motor neurons. To date, no effective treatment or reliable biomarker for ALS has been developed. In recent years, many factors have been proposed as possible biomarkers of ALS; however, no consensus has been reached. Therefore, a reliable biomarker is urgently needed. Eosinophils may play a crucial role in healthy humans and diseases, and serve as a biomarker for many chronic diseases. Methods Routine blood test results were collected from 66 healthy controls and 59 patients with ALS. The percentages and total numbers of each cell population were analyzed, and the correlation between these indicators and patient ALS functional rating scale-revised (ALSFRS-R) score or disease progression rate (ΔFS score) was analyzed. Results Compared to healthy controls, the number of blood leukocytes, neutrophils, monocytes, and basophils was significantly decreased in patients with ALS (p = 0.002, p = 0.001, p = 0.049, and p < 0.0001, respectively). There was an increase in the number of eosinophils (p < 0.0001), but no difference in the number of lymphocytes between patients with ALS and healthy controls was found (p = 0.563). Compared to healthy controls, the percentage of neutrophils was decreased and the percentage of lymphocytes and eosinophils was increased in patients with ALS (p = 0.01, p = 0.012, and p = 0.001, respectively). There was no difference between patients with ALS and healthy controls in the percentage of monocytes and basophils (p = 0.622 and p = 0.09, respectively). However, only the percentage and number of eosinophils had a correlation with the ΔFS score. Further multivariate analysis revealed a significant correlation between the disease duration, eosinophil count and percentage, and the disease progression rate (p < 0.0001, p = 0.048, and p = 0.023, respectively). The neutrophil-to-eosinophil ratio (NER), lymphocyte-to-eosinophil ratio (LER), and monocyte-to-eosinophil ratio (MER) were significantly lower in patients with ALS than in healthy controls. However, only the LER was significantly correlated with the ΔFS score. Conclusion These observations implicate neutrophils, lymphocytes, and eosinophils as important factors, and increasing eosinophil counts were negatively correlated with the ΔFS score in patients with ALS.
Collapse
Affiliation(s)
- Jing Yang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Tingting Liu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Lei Zhang
- Department of Emergency, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xin Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Feng Ping Du
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Qi Liu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Hui Dong
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| | - Yaling Liu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiazhuang, China
- The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, China
- Neurological Laboratory of Hebei Province, Shijiazhuang, China
| |
Collapse
|
22
|
Dubowsky M, Theunissen F, Carr JM, Rogers ML. The Molecular Link Between TDP-43, Endogenous Retroviruses and Inflammatory Neurodegeneration in Amyotrophic Lateral Sclerosis: a Potential Target for Triumeq, an Antiretroviral Therapy. Mol Neurobiol 2023; 60:6330-6345. [PMID: 37450244 PMCID: PMC10533598 DOI: 10.1007/s12035-023-03472-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023]
Abstract
Amyotrophic lateral sclerosis (ALS), also known as motor neuron disease (MND), is a progressive neurological disorder, characterised by the death of upper and lower motor neurons. The aetiology of ALS remains unknown, and treatment options are limited. Endogenous retroviruses (ERVs), specifically human endogenous retrovirus type K (HERV-K), have been proposed to be involved in the propagation of neurodegeneration in ALS. ERVs are genomic remnants of ancient viral infection events, with most being inactive and not retaining the capacity to encode a fully infectious virus. However, some ERVs retain the ability to be activated and transcribed, and ERV transcripts have been found to be elevated within the brain tissue of MND patients. A hallmark of ALS pathology is altered localisation of the transactive response (TAR) DNA binding protein 43 kDa (TDP-43), which is normally found within the nucleus of neuronal and glial cells and is involved in RNA regulation. In ALS, TDP-43 aggregates within the cytoplasm and facilitates neurodegeneration. The involvement of ERVs in ALS pathology is thought to occur through TDP-43 and neuroinflammatory mediators. In this review, the proposed involvement of TDP-43, HERV-K and immune regulators on the onset and progression of ALS will be discussed. Furthermore, the evidence supporting a therapy based on targeting ERVs in ALS will be reviewed.
Collapse
Affiliation(s)
- Megan Dubowsky
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, Australia.
| | - Frances Theunissen
- Perron Institute for Neurological and Translational Science, Nedlands, WA, Australia
- Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Murdoch, WA, Australia
| | - Jillian M Carr
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, Australia
| | - Mary-Louise Rogers
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA, Australia
| |
Collapse
|
23
|
Theme 03 - In Vitro Experimental Models. Amyotroph Lateral Scler Frontotemporal Degener 2023; 24:115-127. [PMID: 37966318 DOI: 10.1080/21678421.2023.2260193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
|
24
|
Chi B, Öztürk MM, Paraggio CL, Leonard CE, Sanita ME, Dastpak M, O’Connell JD, Coady JA, Zhang J, Gygi SP, Lopez-Gonzalez R, Yin S, Reed R. Causal ALS genes impact the MHC class II antigen presentation pathway. Proc Natl Acad Sci U S A 2023; 120:e2305756120. [PMID: 37722062 PMCID: PMC10523463 DOI: 10.1073/pnas.2305756120] [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: 04/10/2023] [Accepted: 08/18/2023] [Indexed: 09/20/2023] Open
Abstract
Mutations in RNA/DNA-binding proteins cause amyotrophic lateral sclerosis (ALS), but the underlying disease mechanisms remain unclear. Here, we report that a set of ALS-associated proteins, namely FUS, EWSR1, TAF15, and MATR3, impact the expression of genes encoding the major histocompatibility complex II (MHC II) antigen presentation pathway. Both subunits of the MHC II heterodimer, HLA-DR, are down-regulated in ALS gene knockouts/knockdown in HeLa and human microglial cells, due to loss of the MHC II transcription factor CIITA. Importantly, hematopoietic progenitor cells (HPCs) derived from human embryonic stem cells bearing the FUSR495X mutation and HPCs derived from C9ORF72 ALS patient induced pluripotent stem cells also exhibit disrupted MHC II expression. Given that HPCs give rise to numerous immune cells, our data raise the possibility that loss of the MHC II pathway results in global failure of the immune system to protect motor neurons from damage that leads to ALS.
Collapse
Affiliation(s)
- Binkai Chi
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Muhammet M. Öztürk
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Christina L. Paraggio
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Claudia E. Leonard
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Maria E. Sanita
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Mahtab Dastpak
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Jeremy D. O’Connell
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Jordan A. Coady
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Jiuchun Zhang
- Harvard Medical School Cell Biology Initiative for Genome Editing and Neurodegeneration, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Steven P. Gygi
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| | - Rodrigo Lopez-Gonzalez
- Department of Neurosciences Lerner Research Institute, Cleveland Clinic, Cleveland, OH44196
| | - Shanye Yin
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY10461
| | - Robin Reed
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA02115
| |
Collapse
|
25
|
Mandrioli J, D'Amico R, Zucchi E, De Biasi S, Banchelli F, Martinelli I, Simonini C, Lo Tartaro D, Vicini R, Fini N, Gianferrari G, Pinti M, Lunetta C, Gerardi F, Tarlarini C, Mazzini L, De Marchi F, Scognamiglio A, Sorarù G, Fortuna A, Lauria G, Bella ED, Caponnetto C, Meo G, Chio A, Calvo A, Cossarizza A. Randomized, double-blind, placebo-controlled trial of rapamycin in amyotrophic lateral sclerosis. Nat Commun 2023; 14:4970. [PMID: 37591957 PMCID: PMC10435464 DOI: 10.1038/s41467-023-40734-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 07/31/2023] [Indexed: 08/19/2023] Open
Abstract
In preclinical studies rapamycin was found to target neuroinflammation, by expanding regulatory T cells, and affecting autophagy, two pillars of amyotrophic lateral sclerosis (ALS) pathogenesis. Herein we report a multicenter, randomized, double-blind trial, in 63 ALS patients who were randomly assigned in a 1:1:1 ratio to receive rapamycin 2 mg/m2/day,1 mg/m2/day or placebo (EUDRACT 2016-002399-28; NCT03359538). The primary outcome, the number of patients exhibiting an increase >30% in regulatory T cells from baseline to treatment end, was not attained. Secondary outcomes were changes from baseline of T, B, NK cell subpopulations, inflammasome mRNA expression and activation status, S6-ribosomal protein phosphorylation, neurofilaments; clinical outcome measures of disease progression; survival; safety and quality of life. Of the secondary outcomes, rapamycin decreased mRNA relative expression of the pro-inflammatory cytokine IL-18, reduced plasmatic IL-18 protein, and increased the percentage of classical monocytes and memory switched B cells, although no corrections were applied for multiple tests. In conclusion, we show that rapamycin treatment is well tolerated and provides reassuring safety findings in ALS patients, but further trials are necessary to understand the biological and clinical effects of this drug in ALS.
Collapse
Affiliation(s)
- Jessica Mandrioli
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.
- Department of Neurosciences, St. Agostino-Estense Hospital, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy.
| | - Roberto D'Amico
- Unit of Statistical and Methodological Support to Clinical Research, Azienda Ospedaliero-Universitaria, Modena, Italy
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Elisabetta Zucchi
- Department of Neurosciences, St. Agostino-Estense Hospital, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
- Neurosciences PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Federico Banchelli
- Unit of Statistical and Methodological Support to Clinical Research, Azienda Ospedaliero-Universitaria, Modena, Italy
| | - Ilaria Martinelli
- Department of Neurosciences, St. Agostino-Estense Hospital, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Cecilia Simonini
- Department of Neurosciences, St. Agostino-Estense Hospital, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberto Vicini
- Unit of Statistical and Methodological Support to Clinical Research, Azienda Ospedaliero-Universitaria, Modena, Italy
| | - Nicola Fini
- Department of Neurosciences, St. Agostino-Estense Hospital, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Giulia Gianferrari
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Department of Neurosciences, St. Agostino-Estense Hospital, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Marcello Pinti
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125, Modena, Italy
| | - Christian Lunetta
- NEuroMuscular Omnicenter, Serena Onlus Foundation, Milan, Italy
- Istituto Maugeri IRCCS Milano, Milan, Italy
| | | | | | - Letizia Mazzini
- ALS Centre, Neurologic Clinic, Maggiore della Carità University Hospital, Novara, Italy
| | - Fabiola De Marchi
- ALS Centre, Neurologic Clinic, Maggiore della Carità University Hospital, Novara, Italy
| | - Ada Scognamiglio
- ALS Centre, Neurologic Clinic, Maggiore della Carità University Hospital, Novara, Italy
| | - Gianni Sorarù
- Department of Neurosciences, University of Padua, Padua, Italy
- Centro Regionale Specializzato Malattie del Motoneurone, Azienda Ospedale Università di Padova, Padua, Italy
| | - Andrea Fortuna
- Department of Neurosciences, University of Padua, Padua, Italy
| | - Giuseppe Lauria
- 3rd Neurology Unit and ALS Centre, IRCCS 'Carlo Besta' Neurological Institute, Milan, Italy
| | - Eleonora Dalla Bella
- 3rd Neurology Unit and ALS Centre, IRCCS 'Carlo Besta' Neurological Institute, Milan, Italy
| | - Claudia Caponnetto
- Department of Neurosciences, Rehabilitatioņ Ophthalmology, Genetics, Mother and Child Disease, Ospedale Policlinico San Martino, Genova, Italy
| | - Giuseppe Meo
- Department of Neurosciences, Rehabilitatioņ Ophthalmology, Genetics, Mother and Child Disease, Ospedale Policlinico San Martino, Genova, Italy
| | - Adriano Chio
- 'Rita Levi Montalcini' Department of Neurosciences, ALS Centre, University of Turin and Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Andrea Calvo
- 'Rita Levi Montalcini' Department of Neurosciences, ALS Centre, University of Turin and Azienda Ospedaliero Universitaria Città della Salute e della Scienza, Turin, Italy
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
- National Institute for Cardiovascular Research, via Irnerio 48, 40126, Bologna, Italy
| |
Collapse
|
26
|
Zheng W, Liang Y, Fan D, He J. Editorial: Common pathogenic mechanism of cerebrovascular disease and degenerative diseases. Front Neurosci 2023; 17:1233204. [PMID: 37425002 PMCID: PMC10327600 DOI: 10.3389/fnins.2023.1233204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 06/13/2023] [Indexed: 07/11/2023] Open
Affiliation(s)
- Wei Zheng
- Department of Neurology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
- Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing, China
| | - Yuqing Liang
- Peking University Health Science Center, Beijing, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
- Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing, China
| | - Ji He
- Department of Neurology, Peking University Third Hospital, Beijing, China
- Beijing Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
- Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing, China
- Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, China
| |
Collapse
|
27
|
Johnson AM, Lukens JR. The innate immune response in tauopathies. Eur J Immunol 2023; 53:e2250266. [PMID: 36932726 PMCID: PMC10247424 DOI: 10.1002/eji.202250266] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/23/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023]
Abstract
Tauopathies, which include frontotemporal dementia, Alzheimer's disease, and chronic traumatic encephalopathy, are a class of neurological disorders resulting from pathogenic tau aggregates. These aggregates disrupt neuronal health and function leading to the cognitive and physical decline of tauopathy patients. Genome-wide association studies and clinical evidence have brought to light the large role of the immune system in inducing and driving tau-mediated pathology. More specifically, innate immune genes are found to harbor tauopathy risk alleles, and innate immune pathways are upregulated throughout the course of disease. Experimental evidence has expanded on these findings by describing pivotal roles for the innate immune system in the regulation of tau kinases and tau aggregates. In this review, we summarize the literature implicating innate immune pathways as drivers of tauopathy.
Collapse
Affiliation(s)
- Alexis M. Johnson
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA 22908, USA
- Neuroscience Graduate Program, UVA, Charlottesville, VA 22908, USA
- BIG Training Graduate Program, UVA, Charlottesville, VA 22908, USA
| | - John R. Lukens
- Center for Brain Immunology and Glia (BIG), Department of Neuroscience, University of Virginia (UVA), Charlottesville, VA 22908, USA
- Neuroscience Graduate Program, UVA, Charlottesville, VA 22908, USA
- BIG Training Graduate Program, UVA, Charlottesville, VA 22908, USA
| |
Collapse
|
28
|
Kurlawala Z, McMillan JD, Singhal RA, Morehouse J, Burke DA, Sears SM, Duregon E, Beverly LJ, Siskind LJ, Friedland RP. Mutant and curli-producing E. coli enhance the disease phenotype in a hSOD1-G93A mouse model of ALS. Sci Rep 2023; 13:5945. [PMID: 37045868 PMCID: PMC10097672 DOI: 10.1038/s41598-023-32594-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 03/29/2023] [Indexed: 04/14/2023] Open
Abstract
The gut microbiome is a potential non-genetic contributing factor for Amyotrophic Lateral Sclerosis. Differences in gut microbial communities have been detected between ALS subjects and healthy controls, including an increase in Escherichia coli in ALS subjects. E. coli and other gram-negative bacteria produce curli proteins, which are functional bacterial amyloids. We examined whether long-term curli overexposure in the gut can exacerbate the development and progression of ALS. We utilized the slow-developing hSOD1-G93A mouse model of ALS with their C57BL/6J WT littermate controls, including males and females, with a total of 91 animals. These mice were on a normal chow diet and fed curli-producing or curli-nonproducing (mutant) E. coli in applesauce (vehicle) 3 times/week, from 1 through 7 months of age. Male hSOD1 mice demonstrated gradual slowing in running speed month 4 onwards, while females exhibited no signs of locomotive impairment even at 7 months of age. Around the same time, male hSOD1 mice showed a gradual increase in frequency of peripheral CD19+ B cells. Among the male hSOD1 group, chronic gut exposure to curli-producing E. coli led to significant shifts in α- and β-diversities. Curli-exposed males showed suppression of immune responses in circulation, but an increase in markers of inflammation, autophagy and protein turnover in skeletal muscle. Some of these markers were also changed in mutant E. coli-exposed mice, including astrogliosis in the brainstem and demyelination in the lumbar spinal cord. Overall, chronic overexposure to a commensal bacteria like E. coli led to distant organ pathology in our model, without the presence of a leaky gut at 6 months. Mechanisms underlying gut-distant organ communication are of tremendous interest to all disciplines.
Collapse
Affiliation(s)
- Zimple Kurlawala
- Department of Neurology, University of Louisville, Louisville, KY, 40202, USA
| | | | - Richa A Singhal
- KY IDeA Networks of Biomedical Research Excellence Bioinformatics Core, University of Louisville, Louisville, KY, 40202, USA
| | - Johnny Morehouse
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, 40202, USA
| | - Darlene A Burke
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, 40202, USA
| | - Sophia M Sears
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA
| | - Eleonora Duregon
- National Institute on Aging, Translational Gerontology, NIH, Bethesda, USA, Maryland
| | - Levi J Beverly
- School of Medicine, University of Louisville, Louisville, KY, 40202, USA
| | - Leah J Siskind
- Department of Pharmacology and Toxicology, University of Louisville, Louisville, KY, 40202, USA
| | - Robert P Friedland
- Department of Neurology, University of Louisville, Louisville, KY, 40202, USA.
| |
Collapse
|
29
|
Evaluation of the Anti-Inflammatory Properties of Mastic Oil Extracted from Pistacia lentiscus var. chia. IMMUNO 2023. [DOI: 10.3390/immuno3010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
Abstract
Mastic oil (MO) is extracted from the resin of the bark of Pistacia lentiscus var. chia, a tree abundantly grown in the Greek island of Chios. Various biological activities, such as antimicrobial, anticancer and antioxidant, have been associated with the dietary intake of MO. However, little is known about MO’s potential anti-inflammatory effects, while some of its main chemical constituents were reported to exert significant anti-inflammatory activity. This study aims to assay the bioactivity of MO on in vitro and in vivo experimental inflammation models, in particular on LPS-stimulated RAW264.7 macrophages, murine primary peritoneal macrophages and a model of zymosan-induced peritonitis in BALB/c mice. The per os administration of MO inhibited the recruitment of macrophages into the peritoneal cavity of zymosan-treated mice, but did not affect neutrophil mobilisation or the levels of IL-6 or TNF-α in the peritoneal fluid. Similarly, IL-6 and TNF-α secretion in primary LPS-stimulated macrophages was not affected by MO, but the levels of phosphoproteins that activate inflammation in macrophages were differentially regulated. Finally, MO and some of its individual constituents reduced nitric oxide (NO), prostaglandin E2 and TNF-α levels in supernatants of LPS-stimulated RAW264.7 cells and inhibited their phagocytosis rate. Our data imply that MO may promote an anti-inflammatory transition in macrophages due to the combined bioactivities of its individual constituents. Thus, as a mixture of various compounds, MO seems to affect multiple molecular mechanisms that are involved in the development of inflammation. Therefore, more research, focusing on MO’s individual constituents and employing various pre-clinical inflammation models that activate different mechanisms, is required for a detailed investigation of the oil’s potential anti-inflammatory activity.
Collapse
|
30
|
Nona RJ, Greer JM, Henderson RD, McCombe PA. HLA and amyotrophic lateral sclerosis: a systematic review and meta-analysis. Amyotroph Lateral Scler Frontotemporal Degener 2023; 24:24-32. [PMID: 35616250 DOI: 10.1080/21678421.2022.2078665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Background: Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease associated with loss of upper and lower motor neurones. It leads to death by respiratory failure and has a typical prognosis of 2-3 years. The immune system has been shown to play a role in the pathophysiology of ALS. Some of the most important immune genes are within the human leukocyte antigen (HLA) region, and a recent genome-wide association study (GWAS) has identified a risk allele for ALS within the HLA region. Older studies have also suggested an HLA association with ALS, with certain HLA alleles showing differing expression between patients and controls. This systematic review and meta-analysis examines the previous studies performed in this field.Methods: We used established publication search engines. Findings were excluded if they did not meet the selection criteria. We then undertook statistical meta-analysis on the eligible papers, using a fixed effects model.Results: There were eight eligible papers. There were three statistically significant meta-analysis findings, although these would not be significant after correction for multiple comparisons. The frequencies of HLA-A9 and HLA-DR4 genotypes were lower in ALS subjects than controls, and HLA-B35 was higher in ALS subjects.Discussion: This systematic review and meta-analysis do not confirm all the previously reported associations of HLA with ALS, but shows three alleles of interest. However, there are limitations to the studies, which include the use of older serotyping methodology and the small numbers of subjects. Given the recent GWAS association with HLA, further modern HLA studies are warranted.
Collapse
Affiliation(s)
- R J Nona
- The University of Queensland Centre for Clinical Research, Brisbane, Australia.,Department of Neurology, the Royal Brisbane & Women's Hospital, Brisbane, Australia
| | - J M Greer
- The University of Queensland Centre for Clinical Research, Brisbane, Australia
| | - R D Henderson
- The University of Queensland Centre for Clinical Research, Brisbane, Australia.,Department of Neurology, the Royal Brisbane & Women's Hospital, Brisbane, Australia
| | - P A McCombe
- The University of Queensland Centre for Clinical Research, Brisbane, Australia.,Department of Neurology, the Royal Brisbane & Women's Hospital, Brisbane, Australia
| |
Collapse
|
31
|
Chen X, Zhou L, Cui C, Sun J. Evolving markers in amyotrophic lateral sclerosis. Adv Clin Chem 2023. [DOI: 10.1016/bs.acc.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
|
32
|
Klose V, Jesse S, Lewerenz J, Kassubek J, Dorst J, Tumani H, Ludolph AC, Roselli F. CSF oligoclonal IgG bands are not associated with ALS progression and prognosis. Front Neurol 2023; 14:1170360. [PMID: 37213901 PMCID: PMC10196068 DOI: 10.3389/fneur.2023.1170360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 04/19/2023] [Indexed: 05/23/2023] Open
Abstract
Introduction Amyotrophic Lateral Sclerosis (ALS) is characterized by progressive motoneuron degeneration through cell autonomous and non-cell autonomous mechanisms; and the involvement of the innate and adaptive immune system has been hypothesized based on human and murine model data. We have explored if B-cell activation and IgG responses, as detected by IgG Oligoclonal bands (OCB) in serum and cerebrospinal fluid, were associated with ALS or with a subgroup of patients with distinct clinical features. Methods IgG OCB were determined in patients affected by ALS (n=457), Alzheimer Disease (n=516), Mild Cognitive Impairment (n=91), Tension-type Headache (n=152) and idiopathic Facial Palsy (n=94). For ALS patients, clinico-demographic and survival data were prospectively collected in the Register Schabia. Results The prevalence of IgG OCB is comparable in ALS and the four neurological cohorts. When the OCB pattern was considered (highlighting either intrathecal or systemic B-cells activation), no effect of OCB pattern on clinic-demographic parameters and overall. ALS patients with intrathecal IgG synthesis (type 2 and 3) were more likely to display infectious, inflammatory or systemic autoimmune conditions. Discussion These data suggest that OCB are not related to ALS pathophysiology but rather are a finding possibly indicative a coincidental infectious or inflammatory comorbidity that merits further investigation.
Collapse
Affiliation(s)
- Veronika Klose
- Department of Neurology, Ulm University, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE)-Ulm, Ulm, Germany
| | - Sarah Jesse
- Department of Neurology, Ulm University, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE)-Ulm, Ulm, Germany
| | - Jan Lewerenz
- Department of Neurology, Ulm University, Ulm, Germany
| | - Jan Kassubek
- Department of Neurology, Ulm University, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE)-Ulm, Ulm, Germany
| | - Johannes Dorst
- Department of Neurology, Ulm University, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE)-Ulm, Ulm, Germany
| | - Hayrettin Tumani
- Department of Neurology, Ulm University, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE)-Ulm, Ulm, Germany
| | - Albert C. Ludolph
- Department of Neurology, Ulm University, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE)-Ulm, Ulm, Germany
- Neurozentrum Ulm, Ulm, Germany
- *Correspondence: Albert C. Ludolph,
| | - Francesco Roselli
- Department of Neurology, Ulm University, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE)-Ulm, Ulm, Germany
- Neurozentrum Ulm, Ulm, Germany
- Francesco Roselli,
| |
Collapse
|
33
|
Meanti R, Bresciani E, Rizzi L, Coco S, Zambelli V, Dimitroulas A, Molteni L, Omeljaniuk RJ, Locatelli V, Torsello A. Potential Applications for Growth Hormone Secretagogues Treatment of Amyotrophic Lateral Sclerosis. Curr Neuropharmacol 2023; 21:2376-2394. [PMID: 36111771 PMCID: PMC10616926 DOI: 10.2174/1570159x20666220915103613] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/18/2022] [Accepted: 08/01/2022] [Indexed: 11/22/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) arises from neuronal death due to complex interactions of genetic, molecular, and environmental factors. Currently, only two drugs, riluzole and edaravone, have been approved to slow the progression of this disease. However, ghrelin and other ligands of the GHS-R1a receptor have demonstrated interesting neuroprotective activities that could be exploited in this pathology. Ghrelin, a 28-amino acid hormone, primarily synthesized and secreted by oxyntic cells in the stomach wall, binds to the pituitary GHS-R1a and stimulates GH secretion; in addition, ghrelin is endowed with multiple extra endocrine bioactivities. Native ghrelin requires esterification with octanoic acid for binding to the GHS-R1a receptor; however, this esterified form is very labile and represents less than 10% of circulating ghrelin. A large number of synthetic compounds, the growth hormone secretagogues (GHS) encompassing short peptides, peptoids, and non-peptidic moieties, are capable of mimicking several biological activities of ghrelin, including stimulation of GH release, appetite, and elevation of blood IGF-I levels. GHS have demonstrated neuroprotective and anticonvulsant effects in experimental models of pathologies both in vitro and in vivo. To illustrate, some GHS, currently under evaluation by regulatory agencies for the treatment of human cachexia, have a good safety profile and are safe for human use. Collectively, evidence suggests that ghrelin and cognate GHS may constitute potential therapies for ALS.
Collapse
Affiliation(s)
- Ramona Meanti
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza, 20900, Italy
| | - Elena Bresciani
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza, 20900, Italy
| | - Laura Rizzi
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza, 20900, Italy
| | - Silvia Coco
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza, 20900, Italy
| | - Vanessa Zambelli
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza, 20900, Italy
| | - Anna Dimitroulas
- Faculty of Health and Medical Sciences, University of Surrey, Stag Hill, Guildford, GU2 7XH, United Kingdom
| | - Laura Molteni
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza, 20900, Italy
| | - Robert J. Omeljaniuk
- Department of Biology, Lakehead University, 955 Oliver Rd, Thunder Bay, Ontario, P7B 5E1, Canada
| | - Vittorio Locatelli
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza, 20900, Italy
| | - Antonio Torsello
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza, 20900, Italy
| |
Collapse
|
34
|
Wei C, Zhu Y, Li S, Chen W, Li C, Jiang S, Xu R. Identification of an immune-related gene prognostic index for predicting prognosis, immunotherapeutic efficacy, and candidate drugs in amyotrophic lateral sclerosis. Front Cell Neurosci 2022; 16:993424. [PMID: 36589282 PMCID: PMC9798295 DOI: 10.3389/fncel.2022.993424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022] Open
Abstract
Rationale and objectives Considering the great insufficiency in the survival prediction and therapy of amyotrophic lateral sclerosis (ALS), it is fundamental to determine an accurate survival prediction for both the clinical practices and the design of treatment trials. Therefore, there is a need for more accurate biomarkers that can be used to identify the subtype of ALS which carries a high risk of progression to guide further treatment. Methods The transcriptome profiles and clinical parameters of a total of 561 ALS patients in this study were analyzed retrospectively by analysis of four public microarray datasets. Based on the results from a series of analyses using bioinformatics and machine learning, immune signatures are able to be used to predict overall survival (OS) and immunotherapeutic response in ALS patients. Apart from other comprehensive analyses, the decision tree and the nomogram, based on the immune signatures, were applied to guide individual risk stratification. In addition, molecular docking methodology was employed to screen potential small molecular to which the immune signatures might response. Results Immune was determined as a major risk factor contributing to OS among various biomarkers of ALS patients. As compared with traditional clinical features, the immune-related gene prognostic index (IRGPI) had a significantly higher capacity for survival prediction. The determination of risk stratification and assessment was optimized by integrating the decision tree and the nomogram. Moreover, the IRGPI may be used to guide preventative immunotherapy for patients at high risks for mortality. The administration of 2MIU IL2 injection in the short-term was likely to be beneficial for the prolongment of survival time, whose dosage should be reduced to 1MIU if the long-term therapy was required. Besides, a useful clinical application for the IRGPI was to screen potential compounds by the structure-based molecular docking methodology. Conclusion Ultimately, the immune-derived signatures in ALS patients were favorable biomarkers for the prediction of survival probabilities and immunotherapeutic responses, and the promotion of drug development.
Collapse
Affiliation(s)
- Caihui Wei
- Department of Neurology, Jiangxi Provincial People’s Hospital, Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Yu Zhu
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Shu Li
- Department of Neurology, Jiangxi Provincial People’s Hospital, Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Wenzhi Chen
- Department of Neurology, Jiangxi Provincial People’s Hospital, Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Cheng Li
- Department of Neurology, Jiangxi Provincial People’s Hospital, Medical College of Nanchang University, Nanchang, Jiangxi, China,Department of Neurology, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China
| | - Shishi Jiang
- Department of Neurology, Jiangxi Provincial People’s Hospital, Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Renshi Xu
- Department of Neurology, Jiangxi Provincial People’s Hospital, Medical College of Nanchang University, Nanchang, Jiangxi, China,Department of Neurology, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi, China,*Correspondence: Renshi Xu, ;
| |
Collapse
|
35
|
Volonté C, Amadio S. Rethinking purinergic concepts and updating the emerging role of P2X7 and P2X4 in amyotrophic lateral sclerosis. Neuropharmacology 2022; 221:109278. [PMID: 36202258 DOI: 10.1016/j.neuropharm.2022.109278] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/08/2022]
Abstract
The topic of the present review regards the ubiquitous and phylogenetically most ancient prototype of intercellular signaling, the one mediated by extracellular nucleosides and nucleotides, bearing a strong influence on pathophysiological processes in the nervous system. Not by chance, purine and pyrimidine molecules are the most prevalent and ubiquitous chemical messengers in the animal and plant kingdoms, operating through a large plethora of purinergic metabolizing enzymes, P1 and P2 receptors, nucleoside and nucleotide channels and transporters. Because ectonucleotidases degrade the agonists of P2 receptors while simultaneously generate the agonists for P1 receptors, and because several agonists, or antagonists, simultaneously bind and activate, or inhibit, more than one receptor subtype, it follows that an all-inclusive "purinergic network" perspective should be better considered when looking at purinergic actions. This becomes particularly crucial during pathological conditions as for instance amyotrophic lateral sclerosis, where the contribution of purinergic signaling has been demonstrated to differ according to each target cell phenotype and stage of disease progression. Here we will present some newly updated results about P2X7 and P2X4 as the most thoroughly investigated P2 receptors in amyotrophic lateral sclerosis, being aware that the comprehension of their actions is still in progress, and that the purinergic rationale for studying this disease must be however wide-ranging and all-inclusive. This article is part of the Special Issue on 'Purinergic Signaling: 50 years'.
Collapse
Affiliation(s)
- Cinzia Volonté
- CNR-Institute for Systems Analysis and Computer Science "Antonio Ruberti", Via Dei Taurini 19, 00185, Rome, Italy; IRCCS Fondazione Santa Lucia-Cellular Neurobiology Unit, Via Del Fosso di Fiorano 65, 00143, Rome, Italy.
| | - Susanna Amadio
- IRCCS Fondazione Santa Lucia-Cellular Neurobiology Unit, Via Del Fosso di Fiorano 65, 00143, Rome, Italy
| |
Collapse
|
36
|
Aydemir D, Surucu S, Basak AN, Ulusu NN. Evaluation of the Hematological and Serum Biochemistry Parameters in the Pre-Symptomatic and Symptomatic Stages of ALS Disease to Support Early Diagnosis and Prognosis. Cells 2022; 11:cells11223569. [PMID: 36428998 PMCID: PMC9688239 DOI: 10.3390/cells11223569] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/03/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is the most common motor neuron disease. Since there are no pathognomonic tests for ALS prognoses; clinical diagnoses of the disease take time and are usually difficult. Prognostic biomarkers are urgently needed for rapid and effective ALS prognoses. Male albino rats were divided into ten groups based on age: 0 (40-45 days old), A (70-75 days old), B (90-95 days old), C (110-115 days old), and D (130-135 days old). Each group was divided into two subgroups according to its mutation status: wild type (SOD1WT) or mutated (SOD1G93A). Serum biochemistry and hematological parameters were measured in 90 rats to evaluate possible biomarkers for faster ALS diagnoses and prognoses. Weight loss, cholesterol, creatinine, glucose, total bilirubin (TBIL), blood urine nitrogen (BUN), c-peptide, glucagon, PYY, white blood cell (WBC), lymphocyte (LYM), monocyte (MID), granulocyte (GRAN), red cell distribution width with standard deviation (RDW-SD), red cell distribution width with the coefficient of variation (RDW-CV), platelet (PLT), mean platelet volume (MPV), platelet distribution width (PDW), and procalcitonin (PCT) levels were changed in the SOD1G93A rats compared to the SOD1WT rats independently from aging. For the first time in the literature, we showed promising hematological and serum biochemistry parameters in the pre-symptomatic and symptomatic stages of ALS by eliminating the effects of aging. Our results can be used for early diagnoses and prognoses of ALS, improving the quality of life and survival time of ALS patients.
Collapse
Affiliation(s)
- Duygu Aydemir
- Department of Medical Biochemistry, School of Medicine, Koc University, Sariyer, Istanbul 34450, Turkey
- Koç University Research Center for Translational Medicine (KUTTAM), Sariyer, Istanbul 34450, Turkey
| | - Selcuk Surucu
- Department of Anatomy, School of Medicine, Koc University, Sariyer, Istanbul 34450, Turkey
| | - Ayse Nazli Basak
- Suna and İnan Kıraç Foundation, Neurodegeneration Research Laboratory, NDAL-KUTTAM, School of Medicine, Koç University, Istanbul 34010, Turkey
| | - Nuriye Nuray Ulusu
- Department of Medical Biochemistry, School of Medicine, Koc University, Sariyer, Istanbul 34450, Turkey
- Koç University Research Center for Translational Medicine (KUTTAM), Sariyer, Istanbul 34450, Turkey
- Correspondence:
| |
Collapse
|
37
|
Gong Z, Liu Y, Ding F, Ba L, Zhang M. Natural killer cells-related immune traits and amyotrophic lateral sclerosis: A Mendelian randomization study. Front Neurosci 2022; 16:981371. [PMID: 36248644 PMCID: PMC9562140 DOI: 10.3389/fnins.2022.981371] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 08/31/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundObservational studies have suggested that peripheral immune disorders are associated with amyotrophic lateral sclerosis (ALS). Previous studies predominantly focused on changes in adaptive immunity. However, emerging evidence showed natural killer (NK) cells, an essential component of innate immunity, were involved in the degeneration of motor neurons. However, the causal relationship between dysregulated NK cells-related immune traits and ALS remains unclear.ObjectiveThis study aimed to explore the causal relationship between NK cells-related immune traits and the risk of ALS.Materials and methodsSingle nucleotide polymorphisms (SNPs) significantly associated with NK cells-related immune traits were selected as instrumental variables to estimate their causal effects on ALS. SNPs from a genome-wide association study (GWAS) on NK cells-related immune traits were used as exposure instruments, including an absolute NK-cells count, absolute HLA-DR+ NK-cells count, NK cells/lymphocytes, NK cells/CD3– lymphocytes, HLA DR+ NK cells/NK cells, HLA DR+ NK cells/CD3– lymphocytes, and the median fluorescence intensities of CD16–CD56+ on NK cells and HLA-DR+ NK cells. Summary-level GWAS statistics of ALS were used as the outcome data. Exposure and outcome data were analyzed using the two-sample Mendelian randomization (MR) method.ResultsEach one standard deviation increase in the expression levels of CD16–CD56+ on NK cells and HLA-DR+ NK cells were associated with a lower risk of ALS in both the MR-Egger and inverse variance weighted methods (P < 0.05). The results proved robust under all sensitivity analyses. Neither instrumental outliers nor heterogeneity were detected.ConclusionOur results suggest that higher expression levels of CD16–CD56+ on NK cells and HLA-DR+ NK cells are associated with a lower risk of ALS.
Collapse
Affiliation(s)
- Zhenxiang Gong
- Department of Neurology and Psychiatry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Liu
- Department of Neurology and Psychiatry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengfei Ding
- Department of Pharmacology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Li Ba
- Department of Neurology and Psychiatry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Li Ba,
| | - Min Zhang
- Department of Neurology and Psychiatry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Li Ba,
| |
Collapse
|
38
|
Elevated Cerebrospinal Fluid Proteins and Albumin Determine a Poor Prognosis for Spinal Amyotrophic Lateral Sclerosis. Int J Mol Sci 2022; 23:ijms231911063. [PMID: 36232365 PMCID: PMC9570498 DOI: 10.3390/ijms231911063] [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: 07/29/2022] [Revised: 09/10/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a heterogeneous disease, both in its onset phenotype and in its rate of progression. The aim of this study was to establish whether the dysfunction of the blood–brain barrier (BBB) and blood–spinal cord barrier (BSCB) measured through cerebrospinal fluid (CSF) proteins and the albumin-quotient (QAlb) are related to the speed of disease progression. An amount of 246 patients diagnosed with ALS were included. CSF and serum samples were determined biochemically for different parameters. Survival analysis based on phenotype shows higher probability of death for bulbar phenotype compared to spinal phenotype (p-value: 0.0006). For the effect of CSF proteins, data shows an increased risk of death for spinal ALS patients as the value of CSF proteins increases. The same model replicated for CSF albumin yielded similar results. Statistical models determined that the lowest cut-off value for CSF proteins able to differentiate patients with a good prognosis and worse prognosis corresponds to CSF proteins ≥ 0.5 g/L (p-value: 0.0189). For the CSF albumin, the QAlb ≥0.65 is associated with elevated probability of death (p-value: 0.0073). High levels of QAlb are a bad prognostic indicator for the spinal phenotype, in addition to high CSF proteins levels that also act as a marker of poor prognosis.
Collapse
|
39
|
Drosophila melanogaster as a Tool for Amyotrophic Lateral Sclerosis Research. J Dev Biol 2022; 10:jdb10030036. [PMID: 36135369 PMCID: PMC9505035 DOI: 10.3390/jdb10030036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/20/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Reliable animal model systems are an integral part of biological research. Ever since Thomas Hunt Morgan won a Nobel Prize for genetic work done using the fruit fly (Drosophila melanogaster) as a model organism, it has played a larger and more important role in genetic research. Drosophila models have long been used to study neurodegenerative diseases and have aided in identifying key disease progression biological pathways. Due to the availability of a vast array of genetic manipulation tools, its relatively short lifespan, and its ability to produce many progenies, D. melanogaster has provided the ability to conduct large-scale genetic screens to elucidate possible genetic and molecular interactions in neurodegenerative diseases such as Alzheimer’s disease, Parkinson’s disease, Huntington’s Disease, and Amyotrophic Lateral Sclerosis (ALS). With regards to ALS, many of the gene mutations that have been discovered to be linked to the disease have been modeled in Drosophila to provide a look into a detailed model of pathogenesis. The aim of this review is to summarize key and newer developments in ALS research that have utilized Drosophila and to provide insight into the profound use of Drosophila as a tool for modeling this disease.
Collapse
|
40
|
Juengling FD, Wuest F, Kalra S, Agosta F, Schirrmacher R, Thiel A, Thaiss W, Müller HP, Kassubek J. Simultaneous PET/MRI: The future gold standard for characterizing motor neuron disease-A clinico-radiological and neuroscientific perspective. Front Neurol 2022; 13:890425. [PMID: 36061999 PMCID: PMC9428135 DOI: 10.3389/fneur.2022.890425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 07/20/2022] [Indexed: 01/18/2023] Open
Abstract
Neuroimaging assessment of motor neuron disease has turned into a cornerstone of its clinical workup. Amyotrophic lateral sclerosis (ALS), as a paradigmatic motor neuron disease, has been extensively studied by advanced neuroimaging methods, including molecular imaging by MRI and PET, furthering finer and more specific details of the cascade of ALS neurodegeneration and symptoms, facilitated by multicentric studies implementing novel methodologies. With an increase in multimodal neuroimaging data on ALS and an exponential improvement in neuroimaging technology, the need for harmonization of protocols and integration of their respective findings into a consistent model becomes mandatory. Integration of multimodal data into a model of a continuing cascade of functional loss also calls for the best attempt to correlate the different molecular imaging measurements as performed at the shortest inter-modality time intervals possible. As outlined in this perspective article, simultaneous PET/MRI, nowadays available at many neuroimaging research sites, offers the perspective of a one-stop shop for reproducible imaging biomarkers on neuronal damage and has the potential to become the new gold standard for characterizing motor neuron disease from the clinico-radiological and neuroscientific perspectives.
Collapse
Affiliation(s)
- Freimut D. Juengling
- Division of Oncologic Imaging, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Faculty of Medicine, University Bern, Bern, Switzerland
| | - Frank Wuest
- Division of Oncologic Imaging, University of Alberta, Edmonton, AB, Canada
| | - Sanjay Kalra
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Neurology, University of Alberta, Edmonton, AB, Canada
| | - Federica Agosta
- Division of Neuroscience, San Raffaele Scientific Institute, University Vita Salute San Raffaele, Milan, Italy
| | - Ralf Schirrmacher
- Division of Oncologic Imaging, University of Alberta, Edmonton, AB, Canada
- Medical Isotope and Cyclotron Facility, University of Alberta, Edmonton, AB, Canada
| | - Alexander Thiel
- Lady Davis Institute for Medical Research, Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Wolfgang Thaiss
- Department of Nuclear Medicine, University of Ulm Medical Center, Ulm, Germany
- Department of Diagnostic and Interventional Radiology, University of Ulm Medical Center, Ulm, Germany
| | - Hans-Peter Müller
- Department of Neurology, Ulm University Medical Center, Ulm, Germany
| | - Jan Kassubek
- Department of Neurology, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
41
|
Reactive oxygen species, the trident of Neptune in the hands of hecate; role in different diseases, signaling pathways, and detection methods. Arch Biochem Biophys 2022; 728:109357. [DOI: 10.1016/j.abb.2022.109357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/29/2022] [Accepted: 07/16/2022] [Indexed: 12/22/2022]
|
42
|
Yu W, He J, Cai X, Yu Z, Zou Z, Fan D. Neuroimmune Crosstalk Between the Peripheral and the Central Immune System in Amyotrophic Lateral Sclerosis. Front Aging Neurosci 2022; 14:890958. [PMID: 35592701 PMCID: PMC9110796 DOI: 10.3389/fnagi.2022.890958] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 04/14/2022] [Indexed: 12/28/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal disease characterized by the degeneration and death of motor neurons. Systemic neuroinflammation contributes to the pathogenesis of ALS. The proinflammatory milieu depends on the continuous crosstalk between the peripheral immune system (PIS) and central immune system (CIS). Central nervous system (CNS) resident immune cells interact with the peripheral immune cells via immune substances. Dysfunctional CNS barriers, including the blood–brain barrier, and blood–spinal cord barrier, accelerate the inflammatory process, leading to a systemic self-destructive cycle. This review focuses on the crosstalk between PIS and CIS in ALS. Firstly, we briefly introduce the cellular compartments of CIS and PIS, respectively, and update some new understanding of changes specifically occurring in ALS. Then, we will review previous studies on the alterations of the CNS barriers, and discuss their crucial role in the crosstalk in ALS. Finally, we will review the moveable compartments of the crosstalk, including cytokines, chemokines, and peripheral immune cells which were found to infiltrate the CNS, highlighting the interaction between PIS and CIS. This review aims to provide new insights into pathogenic mechanisms and innovative therapeutic approaches for ALS.
Collapse
Affiliation(s)
- Weiyi Yu
- Department of Neurology, Peking University Third Hospital, Beijing, China
- Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
- Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing, China
| | - Ji He
- Department of Neurology, Peking University Third Hospital, Beijing, China
- Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
- Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing, China
| | - Xiying Cai
- School of Basic Medical Sciences, Peking University, Beijing, China
| | - Zhou Yu
- Department of Neurology, Peking University Third Hospital, Beijing, China
- Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
- Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing, China
| | - Zhangyu Zou
- Department of Neurology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Dongsheng Fan
- Department of Neurology, Peking University Third Hospital, Beijing, China
- Beijing Municipal Key Laboratory of Biomarker and Translational Research in Neurodegenerative Diseases, Beijing, China
- Key Laboratory for Neuroscience, National Health Commission/Ministry of Education, Peking University, Beijing, China
- *Correspondence: Dongsheng Fan,
| |
Collapse
|
43
|
Zamani A, Walker AK, Rollo B, Ayers KL, Farah R, O'Brien TJ, Wright DK. Impaired glymphatic function in the early stages of disease in a TDP-43 mouse model of amyotrophic lateral sclerosis. Transl Neurodegener 2022; 11:17. [PMID: 35287738 PMCID: PMC8922788 DOI: 10.1186/s40035-022-00291-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/17/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Multiple lines of evidence suggest possible impairment of the glymphatic system in amyotrophic lateral sclerosis (ALS). To investigate this, we used in vivo magnetic resonance imaging (MRI) to assess glymphatic function early in the course of disease in a transgenic mouse with doxycycline (Dox)-controlled expression of cytoplasmic human TDP-43 (hTDP-43ΔNLS), mimicking the key pathology implicated in ALS. METHODS Adult TDP-43 transgenic and littermate monogenic control mice underwent longitudinal multimodal MRI one and three weeks after the cessation of Dox feed, together with weekly rotarod assessments of motor performance. Glymphatic function was assessed using dynamic contrast-enhanced MRI to track the clearance of an MR contrast agent injected into the cisterna magna. RESULTS Compared to their littermate controls, TDP-43 mice exhibited progressive neurodegeneration including that within the primary motor cortex, primary somatosensory cortex and corticospinal tract, significant weight loss including gastrocnemius atrophy, and shortened telomere length. Furthermore, in the presence of this ALS-like phenotype, these mice have significantly disrupted glymphatic function. CONCLUSIONS Although the relationship between glymphatic clearance and ALS disease progression remains to be elucidated, these changes occurred very early in the disease course. This provides initial evidence to suggest that the glymphatic system might be a potential therapeutic target in the treatment of ALS.
Collapse
Affiliation(s)
- Akram Zamani
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Adam K Walker
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, 4072, Australia
| | - Ben Rollo
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Katie L Ayers
- The Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC, 3052, Australia.,Department of Pediatrics, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Raysha Farah
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.,Department of Medicine, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - David K Wright
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, 3004, Australia.
| |
Collapse
|
44
|
Discovery of APL-1030, a Novel, High-Affinity Nanofitin Inhibitor of C3-Mediated Complement Activation. Biomolecules 2022; 12:biom12030432. [PMID: 35327625 PMCID: PMC8946527 DOI: 10.3390/biom12030432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/11/2022] [Accepted: 03/07/2022] [Indexed: 02/05/2023] Open
Abstract
Uncontrolled complement activation contributes to multiple immune pathologies. Although synthetic compstatin derivatives targeting C3 and C3b are robust inhibitors of complement activation, their physicochemical and molecular properties may limit access to specific organs, development of bifunctional moieties, and therapeutic applications requiring transgenic expression. Complement-targeting therapeutics containing only natural amino acids could enable multifunctional pharmacology, gene therapies, and targeted delivery for underserved diseases. A Nanofitin library of hyperthermophilic protein scaffolds was screened using ribosome display for C3/C3b-targeting clones mimicking compstatin pharmacology. APL-1030, a recombinant 64-residue Nanofitin, emerged as the lead candidate. APL-1030 is thermostable, binds C3 (KD, 1.59 nM) and C3b (KD, 1.11 nM), and inhibits complement activation via classical (IC50 = 110.8 nM) and alternative (IC50 = 291.3 nM) pathways in Wieslab assays. Pharmacologic activity (determined by alternative pathway inhibition) was limited to primate species of tested sera. C3b-binding sites of APL-1030 and compstatin were shown to overlap by X-ray crystallography of C3b-bound APL-1030. APL-1030 is a novel, high-affinity inhibitor of primate C3-mediated complement activation developed from natural amino acids on the hyperthermophilic Nanofitin platform. Its properties may support novel drug candidates, enabling bifunctional moieties, gene therapy, and tissue-targeted C3 pharmacologics for diseases with high unmet need.
Collapse
|
45
|
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: 56] [Impact Index Per Article: 18.7] [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
|
46
|
Li C, Zhu Y, Chen W, Li M, Yang M, Shen Z, Zhou Y, Wang L, Wang H, Li S, Ma J, Gong M, Xu R. Circulating NAD+ Metabolism-Derived Genes Unveils Prognostic and Peripheral Immune Infiltration in Amyotrophic Lateral Sclerosis. Front Cell Dev Biol 2022; 10:831273. [PMID: 35155438 PMCID: PMC8831892 DOI: 10.3389/fcell.2022.831273] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/13/2022] [Indexed: 12/12/2022] Open
Abstract
Background: Nicotinamide adenine dinucleotide (NAD+) metabolism has drawn more attention on neurodegeneration research; however, the role in Amyotrophic Lateral Sclerosis (ALS) remains to be fully elucidated. Here, the purpose of this study was to investigate whether the circulating NAD+ metabolic-related gene signature could be identified as a reliable biomarker for ALS survival. Methods: A retrospective analysis of whole blood transcriptional profiles and clinical characteristics of 454 ALS patients was conducted in this study. A series of bioinformatics and machine-learning methods were combined to establish NAD+ metabolic-derived risk score (NPRS) to predict overall survival for ALS patients. The associations of clinical characteristic with NPRS were analyzed and compared. Receiver operating characteristic (ROC) and the calibration curve were utilized to assess the efficacy of prognostic model. Besides, the peripheral immune cell infiltration was assessed in different risk subgroups by applying the CIBERSORT algorithm. Results: Abnormal activation of the NAD+ metabolic pathway occurs in the peripheral blood of ALS patients. Four subtypes with distinct prognosis were constructed based on NAD+ metabolism-related gene expression patterns by using the consensus clustering method. A comparison of the expression profiles of genes related to NAD+ metabolism in different subtypes revealed that the synthase of NAD+ was closely associated with prognosis. Seventeen genes were selected to construct prognostic risk signature by LASSO regression. The NPRS exhibited stronger prognostic capacity compared to traditional clinic-pathological parameters. High NPRS was characterized by NAD+ metabolic exuberant with an unfavorable prognosis. The infiltration levels of several immune cells, such as CD4 naive T cells, CD8 T cells, neutrophils and macrophages, are significantly associated with NPRS. Further clinicopathological analysis revealed that NPRS is more appropriate for predicting the prognostic risk of patients with spinal onset. A prognostic nomogram exhibited more accurate survival prediction compared with other clinicopathological features. Conclusions: In conclusion, it was first proposed that the circulating NAD+ metabolism-derived gene signature is a promising biomarker to predict clinical outcomes, and ultimately facilitating the precise management of patients with ALS.
Collapse
Affiliation(s)
- Cheng Li
- Department of Neurology, Jiangxi Provincial People’s Hospital, Affiliated People’s Hospital of Nanchang University, Nanchang, China
| | - Yu Zhu
- Department of Neurology, Jiangxi Provincial People’s Hospital, Affiliated People’s Hospital of Nanchang University, Nanchang, China
- *Correspondence: Yu Zhu, , ; Renshi Xu, ,
| | - Wenzhi Chen
- Department of Neurology, Jiangxi Provincial People’s Hospital, Affiliated People’s Hospital of Nanchang University, Nanchang, China
| | - Menghua Li
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Mi Yang
- Department of Medical Service, The First Hospital of Nanchang, Affiliated Nanchang Hospital of Sun Yat-sen University, Nanchang, China
| | - Ziyang Shen
- School of Computer and Information Engineering, Jiangxi Agricultural University, Nanchang, China
| | - Yiyi Zhou
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Lulu Wang
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Huan Wang
- Department of Neurology, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Shu Li
- Department of Neurology, Jiangxi Provincial People’s Hospital, Affiliated People’s Hospital of Nanchang University, Nanchang, China
| | - Jiacheng Ma
- School of Aircraft Engineering, Nanchang Hangkong University, Nanchang, China
| | - Mengni Gong
- Medical Examination Center, First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Renshi Xu
- Department of Neurology, Jiangxi Provincial People’s Hospital, Affiliated People’s Hospital of Nanchang University, Nanchang, China
- *Correspondence: Yu Zhu, , ; Renshi Xu, ,
| |
Collapse
|
47
|
Staats KA, Borchelt DR, Tansey MG, Wymer J. Blood-based biomarkers of inflammation in amyotrophic lateral sclerosis. Mol Neurodegener 2022; 17:11. [PMID: 35073950 PMCID: PMC8785449 DOI: 10.1186/s13024-022-00515-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 12/30/2021] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS) is a devastating neurodegenerative disease in which many processes are detected including (neuro)inflammation. Many drugs have been tested for ALS in clinical trials but most have failed to reach their primary endpoints. The development and inclusion of different types of biomarkers in diagnosis and clinical trials can assist in determining target engagement of a drug, in distinguishing between ALS and other diseases, and in predicting disease progression rate, drug responsiveness, or an adverse event. Ideally, among other characteristics, a biomarker in ALS correlates highly with a disease process in the central nervous system or with disease progression and is conveniently obtained in a peripheral tissue. Here, we describe the state of biomarkers of inflammation in ALS by focusing on peripherally detectable and cellular responses from blood cells, and provide new (combinatorial) directions for exploration that are now feasible due to technological advancements.
Collapse
Affiliation(s)
- Kim A. Staats
- Staats Life Sciences Consulting, LLC, Los Angeles, CA USA
| | - David R. Borchelt
- Department of Neuroscience, University of Florida College of Medicine, McKnight Brain Institute, Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, Florida USA
| | - Malú Gámez Tansey
- Department of Neuroscience and Center for Translational Research in Neurodegenerative Disease at The University of Florida College of Medicine, Gainesville, Florida USA
| | - James Wymer
- Department of Neurology, University of Florida College of Medicine, Gainesville, Florida USA
| |
Collapse
|
48
|
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder characterized by irreversible deterioration of upper and lower motor neurons (MNs). Previously, studies on the involvement of glial cells in the pathogenic process of ALS have mainly revolved around astrocytes and microglia. And oligodendrocytes (OLs) have only recently been highlighted. Grey matter demyelination within the motor cortex and proliferation of the oligodendrocyte precursor cells (OPCs) was observed in ALS patients. The selective ablation of mutant SOD1 (the dysfunctional superoxide dismutase) from the oligodendrocyte progenitors after birth significantly delayed disease onset and prolonged the overall survival in ALS mice model (SOD1G37R). In this study, we review the several mechanisms of oligodendrocyte dysfunction involved in the pathological process of myelin damage and MNs death during ALS. Particularly, we examined the insufficient local energy supply from OLs to axons, impaired differentiation from OPCs into OLs mediated by oxidative stress damage, and inflammatory injury to the OLs. Since increasing evidence depicted that ALS is not a disease limited to MNs damage, exploring the mechanisms by which oligodendrocyte dysfunction is involved in MNs death would contribute to a more comprehensive understanding of ALS and identifying potential drug targets.
Collapse
Affiliation(s)
- Zhenxiang Gong
- Department of Neurology and Psychiatry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Li Ba
- Department of Neurology and Psychiatry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Min Zhang
- Department of Neurology and Psychiatry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
- Min Zhang, Department of Neurology and Psychiatry, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Road, Qiaokou District, Wuhan, Hubei 430030, China. Tel: +86-27-83663895, E-mail:
| |
Collapse
|
49
|
Contingent intramuscular boosting of P2XR7 axis improves motor function in transgenic ALS mice. Cell Mol Life Sci 2021; 79:7. [PMID: 34936028 PMCID: PMC8695421 DOI: 10.1007/s00018-021-04070-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 11/06/2022]
Abstract
Amyotrophic lateral sclerosis is a fatal neurodegenerative disorder that leads to progressive degeneration of motor neurons and severe muscle atrophy without effective treatment. Most research on the disease has been focused on studying motor neurons and supporting cells of the central nervous system. Strikingly, the recent observations have suggested that morpho-functional alterations in skeletal muscle precede motor neuron degeneration, bolstering the interest in studying muscle tissue as a potential target for the delivery of therapies. We previously showed that the systemic administration of the P2XR7 agonist, 2′(3′)-O‐(4-benzoylbenzoyl) adenosine 5-triphosphate (BzATP), enhanced the metabolism and promoted the myogenesis of new fibres in the skeletal muscles of SOD1G93A mice. Here we further corroborated this evidence showing that intramuscular administration of BzATP improved the motor performance of ALS mice by enhancing satellite cells and the muscle pro-regenerative activity of infiltrating macrophages. The preservation of the skeletal muscle retrogradely propagated along with the motor unit, suggesting that backward signalling from the muscle could impinge on motor neuron death. In addition to providing the basis for a suitable adjunct multisystem therapeutic approach in ALS, these data point out that the muscle should be at the centre of ALS research as a target tissue to address novel therapies in combination with those oriented to the CNS.
Collapse
|
50
|
Frausto DM, Forsyth CB, Keshavarzian A, Voigt RM. Dietary Regulation of Gut-Brain Axis in Alzheimer's Disease: Importance of Microbiota Metabolites. Front Neurosci 2021; 15:736814. [PMID: 34867153 PMCID: PMC8639879 DOI: 10.3389/fnins.2021.736814] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/18/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that impacts 45 million people worldwide and is ranked as the 6th top cause of death among all adults by the Centers for Disease Control and Prevention. While genetics is an important risk factor for the development of AD, environment and lifestyle are also contributing risk factors. One such environmental factor is diet, which has emerged as a key influencer of AD development/progression as well as cognition. Diets containing large quantities of saturated/trans-fats, refined carbohydrates, limited intake of fiber, and alcohol are associated with cognitive dysfunction while conversely diets low in saturated/trans-fats (i.e., bad fats), high mono/polyunsaturated fats (i.e., good fats), high in fiber and polyphenols are associated with better cognitive function and memory in both humans and animal models. Mechanistically, this could be the direct consequence of dietary components (lipids, vitamins, polyphenols) on the brain, but other mechanisms are also likely to be important. Diet is considered to be the single greatest factor influencing the intestinal microbiome. Diet robustly influences the types and function of micro-organisms (called microbiota) that reside in the gastrointestinal tract. Availability of different types of nutrients (from the diet) will favor or disfavor the abundance and function of certain groups of microbiota. Microbiota are highly metabolically active and produce many metabolites and other factors that can affect the brain including cognition and the development and clinical progression of AD. This review summarizes data to support a model in which microbiota metabolites influence brain function and AD.
Collapse
Affiliation(s)
- Dulce M. Frausto
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
| | - Christopher B. Forsyth
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| | - Ali Keshavarzian
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
- Department of Physiology, Rush University Medical Center, Chicago, IL, United States
| | - Robin M. Voigt
- Rush Medical College, Rush Center for Integrated Microbiome and Chronobiology Research, Rush University Medical Center, Chicago, IL, United States
- Department of Medicine, Rush University Medical Center, Chicago, IL, United States
| |
Collapse
|